1
|
Grobben Y. Targeting amino acid-metabolizing enzymes for cancer immunotherapy. Front Immunol 2024; 15:1440269. [PMID: 39211039 PMCID: PMC11359565 DOI: 10.3389/fimmu.2024.1440269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Despite the immune system's role in the detection and eradication of abnormal cells, cancer cells often evade elimination by exploitation of various immune escape mechanisms. Among these mechanisms is the ability of cancer cells to upregulate amino acid-metabolizing enzymes, or to induce these enzymes in tumor-infiltrating immunosuppressive cells. Amino acids are fundamental cellular nutrients required for a variety of physiological processes, and their inadequacy can severely impact immune cell function. Amino acid-derived metabolites can additionally dampen the anti-tumor immune response by means of their immunosuppressive activities, whilst some can also promote tumor growth directly. Based on their evident role in tumor immune escape, the amino acid-metabolizing enzymes glutaminase 1 (GLS1), arginase 1 (ARG1), inducible nitric oxide synthase (iNOS), indoleamine 2,3-dioxygenase 1 (IDO1), tryptophan 2,3-dioxygenase (TDO) and interleukin 4 induced 1 (IL4I1) each serve as a promising target for immunotherapeutic intervention. This review summarizes and discusses the involvement of these enzymes in cancer, their effect on the anti-tumor immune response and the recent progress made in the preclinical and clinical evaluation of inhibitors targeting these enzymes.
Collapse
|
2
|
Antonatos C, Georgakilas GK, Evangelou E, Vasilopoulos Y. Transcriptomic meta-analysis characterizes molecular commonalities between psoriasis and obesity. Genes Immun 2024; 25:179-187. [PMID: 38580831 DOI: 10.1038/s41435-024-00271-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
Despite the abundance of epidemiological evidence for the high comorbid rate between psoriasis and obesity, systematic approaches to common inflammatory mechanisms have not been adequately explored. We performed a meta-analysis of publicly available RNA-sequencing datasets to unveil putative mechanisms that are postulated to exacerbate both diseases, utilizing both late-stage, disease-specific meta-analyses and consensus gene co-expression network (cWGCNA). Single-gene meta-analyses reported several common inflammatory mechanisms fostered by the perturbed expression profile of inflammatory cells. Assessment of gene overlaps between both diseases revealed significant overlaps between up- (n = 170, P value = 6.07 × 10-65) and down-regulated (n = 49, P value = 7.1 × 10-7) genes, associated with increased T cell response and activated transcription factors. Our cWGCNA approach disentangled 48 consensus modules, associated with either the differentiation of leukocytes or metabolic pathways with similar correlation signals in both diseases. Notably, all our analyses confirmed the association of the perturbed T helper (Th)17 differentiation pathway in both diseases. Our novel findings through whole transcriptomic analyses characterize the inflammatory commonalities between psoriasis and obesity implying the assessment of several expression profiles that could serve as putative comorbid disease progression biomarkers and therapeutic interventions.
Collapse
Affiliation(s)
- Charalabos Antonatos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece
| | - Georgios K Georgakilas
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece
- Information Management Systems Institute (IMSI), ATHENA Research Center, 15125, Athens, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110, Ioannina, Greece
- Department of Epidemiology & Biostatistics, MRC Centre for Environment and Health, Imperial College London, London, W2 1PG, UK
| | - Yiannis Vasilopoulos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504, Patras, Greece.
| |
Collapse
|
3
|
Chen B, Liu Y, He Y, Shen C. Pan-cancer analysis of prognostic and immunological role of IL4I1 in human tumors: a bulk omics research and single cell sequencing validation. Discov Oncol 2024; 15:139. [PMID: 38691253 PMCID: PMC11063023 DOI: 10.1007/s12672-024-01000-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 04/29/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Interleukin-4 inducible gene 1 (IL4I1) regulates tumor progression in numerous tumor types. However, its correlation with immune infiltration and prognosis of patients in a pan-cancer setting remains unclear. METHODS Data from the Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), UALCAN, Clinical Proteomic Tumor Analysis Consortium (CPTAC), Gene Expression Omnibus (GEO), cBioPortal, Cancer Single-cell State Atlas (CancerSEA), and Tumor IMmune Estimation Resource(TIMER) databases were used to evaluate IL4I1 expression, clinical features and prognostic effects, gene set enrichment, and correlation with immune cell infiltration, as well as the relationship between IL4I1 methylation and expression and survival prognosis. Correlations with 192 anticancer drugs were also analyzed. RESULTS IL4I1 was significantly overexpressed in the majority of tumors, and the imbalance of IL4I1 was significantly correlated with overall survival and pathological stage. Moreover, total IL4I1 protein was increased in cancer. Therefore, IL4I1 may be used as a prognostic biomarker or protective factor in numerous types of cancer. The methylation level of IL4I1 may also be used as a prognostic marker. The functional enrichment of IL4I1 was closely related to the immunomodulatory pathway. In addition, the level of tumor-associated macrophage infiltration was positively correlated with the expression of IL4I1 in pan-cancerous tissues. scRNA-seq analysis suggested that IL4I1 differ significantly among different cells in the tumor microenvironment and was most enriched in macrophages. Various immune checkpoint genes were positively correlated with IL4I1 expression in most tumors. In addition, patients with high IL4I1 expression may be resistant to BMS-754807 and docetaxel, but sensitive to temozolomide. CONCLUSION IL4I1 may play a role as promoter of cancer and prognostic indicator in patients. High expression of IL4I1 is associated with the state of tumor immunosuppression and may contribute to tumor-associated macrophage invasion. Therefore, IL4I1 may be a new therapeutic target for the treatment and prognosis of patients with cancer.
Collapse
Affiliation(s)
- Bin Chen
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Liu
- Emergency Department, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuping He
- Health Management Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chenfu Shen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Burris TP, de Vera IMS, Cote I, Flaveny CA, Wanninayake US, Chatterjee A, Walker JK, Steinauer N, Zhang J, Coons LA, Korach KS, Cain DW, Hollenberg AN, Webb P, Forrest D, Jetten AM, Edwards DP, Grimm SL, Hartig S, Lange CA, Richer JK, Sartorius CA, Tetel M, Billon C, Elgendy B, Hegazy L, Griffett K, Peinetti N, Burnstein KL, Hughes TS, Sitaula S, Stayrook KR, Culver A, Murray MH, Finck BN, Cidlowski JA. International Union of Basic and Clinical Pharmacology CXIII: Nuclear Receptor Superfamily-Update 2023. Pharmacol Rev 2023; 75:1233-1318. [PMID: 37586884 PMCID: PMC10595025 DOI: 10.1124/pharmrev.121.000436] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
The NR superfamily comprises 48 transcription factors in humans that control a plethora of gene network programs involved in a wide range of physiologic processes. This review will summarize and discuss recent progress in NR biology and drug development derived from integrating various approaches, including biophysical techniques, structural studies, and translational investigation. We also highlight how defective NR signaling results in various diseases and disorders and how NRs can be targeted for therapeutic intervention via modulation via binding to synthetic lipophilic ligands. Furthermore, we also review recent studies that improved our understanding of NR structure and signaling. SIGNIFICANCE STATEMENT: Nuclear receptors (NRs) are ligand-regulated transcription factors that are critical regulators of myriad physiological processes. NRs serve as receptors for an array of drugs, and in this review, we provide an update on recent research into the roles of these drug targets.
Collapse
Affiliation(s)
- Thomas P Burris
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Ian Mitchelle S de Vera
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Isabelle Cote
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Colin A Flaveny
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Udayanga S Wanninayake
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Arindam Chatterjee
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - John K Walker
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Nickolas Steinauer
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Jinsong Zhang
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Laurel A Coons
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Kenneth S Korach
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Derek W Cain
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Anthony N Hollenberg
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Paul Webb
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Douglas Forrest
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Anton M Jetten
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Dean P Edwards
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Sandra L Grimm
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Sean Hartig
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Carol A Lange
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Jennifer K Richer
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Carol A Sartorius
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Marc Tetel
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Cyrielle Billon
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Bahaa Elgendy
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Lamees Hegazy
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Kristine Griffett
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Nahuel Peinetti
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Kerry L Burnstein
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Travis S Hughes
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Sadichha Sitaula
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Keitch R Stayrook
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Alexander Culver
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Meghan H Murray
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - Brian N Finck
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| | - John A Cidlowski
- University of Florida Genetics Institute, Gainesville, Florida (T.P.B., I.C.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (I.M.S.d.V., U.S.W., A.C., J.K.W., N.S., J.Z.); Pfizer, San Diego, California (C.A.F.); Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (L.A.C., K.S.K.); Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina (L.A.C.); Duke Human Vaccine Institute, Durham, North Carolina (D.W.C.); Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (A.N.H.); The California Institute of Regenerative Medicine, South San Francisco, California (P.W.); Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.G.); National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina (A.M.J.); Department of Molecular and Cellular Pharmacology, Baylor College of Medicine, Houston, Texas (D.P.E., S.L.G., S.H.); Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota (C.A.L.); Department of Pathology, University of Colorado, Aurora, Colorado (J.K.R., C.A.S.); Neuroscience Program, Wellesley College, Wellesley, Massachusetts (M.T.); Center for Clinical Pharmacology, University of Health Sciences and Pharmacy, Saint Louis, Missouri (C.B., B.E., L.H.); Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama (K.G.); Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida (N.P., K.L.B.); Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana (T.S.H.); Asteroid Therapeutics, Inc. Indianapolis, Indiana (S.S., K.R.S., A.C.); Saint Louis University School of Medicine, St. Louis, Missouri (M.H.M.); Department of Medicine, Washington University School of Medicine, St. Louis, Missouri (B.N.F.); and Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina (J.A.C.)
| |
Collapse
|
5
|
Sato W, Noto D, Araki M, Okamoto T, Lin Y, Yamaguchi H, Kadowaki-Saga R, Kimura A, Kimura Y, Sato N, Ishizuka T, Nakamura H, Miyake S, Yamamura T. First-in-human clinical trial of the NKT cell-stimulatory glycolipid OCH in multiple sclerosis. Ther Adv Neurol Disord 2023; 16:17562864231162153. [PMID: 36993937 PMCID: PMC10041592 DOI: 10.1177/17562864231162153] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/20/2023] [Indexed: 03/31/2023] Open
Abstract
Background Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system that causes the damage to the myelin sheath as well as axonal degeneration. Individuals with MS appear to have changes in the numbers and functions of T-cell subsets, leading to an immunological imbalance accompanied by enhanced autoreactivity. In previous preclinical studies, (2 S,3 S,4R)-1-O-(α-D-Galactopyranosyl)-N-tetracosanoyl-2-amino-1,3,4-nonanetriol (OCH), a synthetic analog of α-galactosylceramide stimulatory for invariant NKT (iNKT) cells, has shown therapeutic or disease-preventive immunoregulatory effects in autoimmune disease models such as experimental autoimmune encephalomyelitis (EAE). Objectives This study is the first-in-human study of oral OCH to evaluate the pharmacokinetics and to examine the effects on immune cells as well as related gene expression profiles. Methods Fifteen healthy volunteers and 13 MS patients who met the study criteria were enrolled. They were divided into five cohorts and received oral administration of various doses of granulated powder of OCH (0.3-30 mg), once per week for 4 or 13 weeks. Plasma OCH concentrations were measured by high-performance liquid chromatography. Frequencies of lymphocyte subsets in peripheral blood were evaluated by flow cytometry, and microarray analysis was performed to determine OCH-induced changes in gene expression. Results Oral OCH was well tolerated, and its bioavailability was found to be sufficient. Six hours after a single dose of OCH, increased frequencies of Foxp3+ regulatory T-cells were observed in some cohorts of healthy subjects and MS patients. Furthermore, gene expression analysis demonstrated an upregulation of several immunoregulatory genes and downregulation of pro-inflammatory genes following OCH administration. Conclusion This study has demonstrated immunomodulatory effects of the iNKT cell-stimulatory drug OCH in human. Safety profiles together with the presumed anti-inflammatory effects of oral OCH encouraged us to conduct a phase II trial.
Collapse
Affiliation(s)
| | | | - Manabu Araki
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Tomoko Okamoto
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Youwei Lin
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Hiromi Yamaguchi
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Ryoko Kadowaki-Saga
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Atsuko Kimura
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Yukio Kimura
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Noriko Sato
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Takami Ishizuka
- Translational Medical Center, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Harumasa Nakamura
- Translational Medical Center, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | | |
Collapse
|
6
|
Maggi E, Parronchi P, Azzarone BG, Moretta L. A pathogenic integrated view explaining the different endotypes of asthma and allergic disorders. Allergy 2022; 77:3267-3292. [PMID: 35842745 DOI: 10.1111/all.15445] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
The inflammation of allergic diseases is characterized by a complex interaction between type 2 and type 3 immune responses, explaining clinical symptoms and histopathological patterns. Airborne stimuli activate the mucosal epithelium to release a number of molecules impacting the activity of resident immune and environmental cells. Signals from the mucosal barrier, regulatory cells, and the inflamed tissue are crucial conditions able to modify innate and adaptive effector cells providing the selective homing of eosinophils or neutrophils. The high plasticity of resident T- and innate lymphoid cells responding to external signals is the prerequisite to explain the multiplicity of endotypes of allergic diseases. This notion paved the way for the huge use of specific biologic drugs interfering with pathogenic mechanisms of inflammation. Based on the response of the epithelial barrier, the activity of resident regulatory cells, and functions of structural non-lymphoid environmental cells, this review proposes some immunopathogenic scenarios characterizing the principal endotypes which can be associated with a precise phenotype of asthma. Recent literature indicates that similar concepts can also be applied to the inflammation of other non-respiratory allergic disorders. The next challenges will consist in defining specific biomarker(s) of each endotype allowing for a quick diagnosis and the most effective personalized therapy.
Collapse
Affiliation(s)
- Enrico Maggi
- Department of Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paola Parronchi
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | | | - Lorenzo Moretta
- Department of Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
7
|
Hamoudi C, Zhao C, Abderrazak A, Salem M, Fortin PR, Sévigny J, Aoudjit F. The Purinergic Receptor P2X4 Promotes Th17 Activation and the Development of Arthritis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1115-1127. [PMID: 35165166 DOI: 10.4049/jimmunol.2100550] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/28/2021] [Indexed: 01/24/2023]
Abstract
Purinergic signaling plays a major role in T cell activation leading to IL-2 production and proliferation. However, it is unclear whether purinergic signaling contributes to the differentiation and activation of effector T cells. In this study, we found that the purinergic receptor P2X4 was associated with human Th17 cells but not with Th1 cells. Inhibition of P2X4 receptor with the specific antagonist 5-BDBD and small interfering RNA inhibited the development of Th17 cells and the production of IL-17 by effector Th17 cells stimulated via the CD3/CD28 pathway. Our results showed that P2X4 was required for the expression of retinoic acid-related orphan receptor C, which is the master regulator of Th17 cells. In contrast, inhibition of P2X4 receptor had no effect on Th1 cells and on the production of IFN-γ and it did not affect the expression of the transcription factor T-bet (T-box transcription factor). Furthermore, inhibition of P2X4 receptor reduced the production of IL-17 but not of IFN-γ by effector/memory CD4+ T cells isolated from patients with rheumatoid arthritis. In contrast to P2X4, inhibition of P2X7 and P2Y11 receptors had no effects on Th17 and Th1 cell activation. Finally, treatment with the P2X4 receptor antagonist 5-BDBD reduced the severity of collagen-induced arthritis in mice by inhibiting Th17 cell expansion and activation. Our findings provide novel insights into the role of purinergic signaling in T cell activation and identify a critical role for the purinergic receptor P2X4 in Th17 activation and in autoimmune arthritis.
Collapse
Affiliation(s)
- Chakib Hamoudi
- Division of Immune and Infectious Diseases, CHU de Québec Research Center, Quebec City, Quebec, Canada.,ARThrite Center, Laval University, Quebec City, Quebec, Canada
| | - Chenqi Zhao
- Division of Immune and Infectious Diseases, CHU de Québec Research Center, Quebec City, Quebec, Canada.,ARThrite Center, Laval University, Quebec City, Quebec, Canada
| | - Amna Abderrazak
- Division of Immune and Infectious Diseases, CHU de Québec Research Center, Quebec City, Quebec, Canada
| | - Mabrouka Salem
- Division of Immune and Infectious Diseases, CHU de Québec Research Center, Quebec City, Quebec, Canada.,ARThrite Center, Laval University, Quebec City, Quebec, Canada
| | - Paul R Fortin
- Division of Immune and Infectious Diseases, CHU de Québec Research Center, Quebec City, Quebec, Canada.,ARThrite Center, Laval University, Quebec City, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada; and
| | - Jean Sévigny
- Division of Immune and Infectious Diseases, CHU de Québec Research Center, Quebec City, Quebec, Canada.,ARThrite Center, Laval University, Quebec City, Quebec, Canada.,Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
| | - Fawzi Aoudjit
- Division of Immune and Infectious Diseases, CHU de Québec Research Center, Quebec City, Quebec, Canada; .,ARThrite Center, Laval University, Quebec City, Quebec, Canada.,Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
8
|
Rannug A. 6-Formylindolo[3,2-b]carbazole, a Potent Ligand for the Aryl Hydrocarbon Receptor Produced Both Endogenously and by Microorganisms, can Either Promote or Restrain Inflammatory Responses. FRONTIERS IN TOXICOLOGY 2022; 4:775010. [PMID: 35295226 PMCID: PMC8915874 DOI: 10.3389/ftox.2022.775010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) binds major physiological modifiers of the immune system. The endogenous 6-formylindolo[3,2-b]carbazole (FICZ), which binds with higher affinity than any other compound yet tested, including TCDD, plays a well-documented role in maintaining the homeostasis of the intestines and skin. The effects of transient activation of AHR by FICZ differ from those associated with continuous stimulation and, depending on the dose, include either differentiation into T helper 17 cells that express proinflammatory cytokines or into regulatory T cells or macrophages with anti-inflammatory properties. Moreover, in experimental models of human diseases high doses stimulate the production of immunosuppressive cytokines and suppress pathogenic autoimmunity. In our earlier studies we characterized the formation of FICZ from tryptophan via the precursor molecules indole-3-pyruvate and indole-3-acetaldehyde. In the gut formation of these precursor molecules is catalyzed by microbial aromatic-amino-acid transaminase ArAT. Interestingly, tryptophan can also be converted into indole-3-pyruvate by the amino-acid catabolizing enzyme interleukin-4 induced gene 1 (IL4I1), which is secreted by host immune cells. By thus generating derivatives of tryptophan that activate AHR, IL4I1 may have a role to play in anti-inflammatory responses, as well as in a tumor escape mechanism that reduces survival in cancer patients. The realization that FICZ can be produced from tryptophan by sunlight, by enzymes expressed in our cells (IL4I1), and by microorganisms as well makes it highly likely that this compound is ubiquitous in humans. A diurnal oscillation in the level of FICZ that depends on the production by the fluctuating number of microbes might influence not only intestinal and dermal immunity locally, but also systemic immunity.
Collapse
|
9
|
Wang X, Guan S, Sun L, Dai Z. The impact of benzo[a]pyrene on murine allergic airway inflammation via epigenetic remodeling. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 89:103782. [PMID: 34883242 DOI: 10.1016/j.etap.2021.103782] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 06/13/2023]
Abstract
Simultaneous exposure to both BaP and house dust mites (HDM) has been shown to exacerbate pulmonary inflammation and hyperresponsiveness in a murine asthma model. The mechanistic insight into epigenetic inheritance for this effect, however, remains to be clarified. As such, in this study, we explore the molecular basis for the enhancement of asthma. Female BAL/C mice were intranasally administered HDM (25 µg in 25 μL saline) and/or BaP (10 μg/kg) every other day for 9 weeks. RNA sequencing and DNA methylation assessment were used to explore the underlying mechanism. Following simultaneous exposure to HDM and BaP, mice exhibited pulmonary inflammation and the transcript level of IL4i1b, muc4 and IL22ra2 that were associated with altered DNA methylation, suggesting that there may be an epigenetic basis for BaP-induced asthma exacerbation. Our data suggest that DNA methylation is a major epigenetic modification that accompanies airway remodeling associated with changes in the allergic mice.
Collapse
Affiliation(s)
- Xihua Wang
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shuyuan Guan
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lingbin Sun
- The Department of Anesthesiology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, China; Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Zhongliang Dai
- The Department of Anesthesiology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, China.
| |
Collapse
|
10
|
Cossarizza A, Chang HD, Radbruch A, Abrignani S, Addo R, Akdis M, Andrä I, Andreata F, Annunziato F, Arranz E, Bacher P, Bari S, Barnaba V, Barros-Martins J, Baumjohann D, Beccaria CG, Bernardo D, Boardman DA, Borger J, Böttcher C, Brockmann L, Burns M, Busch DH, Cameron G, Cammarata I, Cassotta A, Chang Y, Chirdo FG, Christakou E, Čičin-Šain L, Cook L, Corbett AJ, Cornelis R, Cosmi L, Davey MS, De Biasi S, De Simone G, del Zotto G, Delacher M, Di Rosa F, Di Santo J, Diefenbach A, Dong J, Dörner T, Dress RJ, Dutertre CA, Eckle SBG, Eede P, Evrard M, Falk CS, Feuerer M, Fillatreau S, Fiz-Lopez A, Follo M, Foulds GA, Fröbel J, Gagliani N, Galletti G, Gangaev A, Garbi N, Garrote JA, Geginat J, Gherardin NA, Gibellini L, Ginhoux F, Godfrey DI, Gruarin P, Haftmann C, Hansmann L, Harpur CM, Hayday AC, Heine G, Hernández DC, Herrmann M, Hoelsken O, Huang Q, Huber S, Huber JE, Huehn J, Hundemer M, Hwang WYK, Iannacone M, Ivison SM, Jäck HM, Jani PK, Keller B, Kessler N, Ketelaars S, Knop L, Knopf J, Koay HF, Kobow K, Kriegsmann K, Kristyanto H, Krueger A, Kuehne JF, Kunze-Schumacher H, Kvistborg P, Kwok I, Latorre D, Lenz D, Levings MK, Lino AC, Liotta F, Long HM, Lugli E, MacDonald KN, Maggi L, Maini MK, Mair F, Manta C, Manz RA, Mashreghi MF, Mazzoni A, McCluskey J, Mei HE, Melchers F, Melzer S, Mielenz D, Monin L, Moretta L, Multhoff G, Muñoz LE, Muñoz-Ruiz M, Muscate F, Natalini A, Neumann K, Ng LG, Niedobitek A, Niemz J, Almeida LN, Notarbartolo S, Ostendorf L, Pallett LJ, Patel AA, Percin GI, Peruzzi G, Pinti M, Pockley AG, Pracht K, Prinz I, Pujol-Autonell I, Pulvirenti N, Quatrini L, Quinn KM, Radbruch H, Rhys H, Rodrigo MB, Romagnani C, Saggau C, Sakaguchi S, Sallusto F, Sanderink L, Sandrock I, Schauer C, Scheffold A, Scherer HU, Schiemann M, Schildberg FA, Schober K, Schoen J, Schuh W, Schüler T, Schulz AR, Schulz S, Schulze J, Simonetti S, Singh J, Sitnik KM, Stark R, Starossom S, Stehle C, Szelinski F, Tan L, Tarnok A, Tornack J, Tree TIM, van Beek JJP, van de Veen W, van Gisbergen K, Vasco C, Verheyden NA, von Borstel A, Ward-Hartstonge KA, Warnatz K, Waskow C, Wiedemann A, Wilharm A, Wing J, Wirz O, Wittner J, Yang JHM, Yang J. Guidelines for the use of flow cytometry and cell sorting in immunological studies (third edition). Eur J Immunol 2021; 51:2708-3145. [PMID: 34910301 PMCID: PMC11115438 DOI: 10.1002/eji.202170126] [Citation(s) in RCA: 218] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The third edition of Flow Cytometry Guidelines provides the key aspects to consider when performing flow cytometry experiments and includes comprehensive sections describing phenotypes and functional assays of all major human and murine immune cell subsets. Notably, the Guidelines contain helpful tables highlighting phenotypes and key differences between human and murine cells. Another useful feature of this edition is the flow cytometry analysis of clinical samples with examples of flow cytometry applications in the context of autoimmune diseases, cancers as well as acute and chronic infectious diseases. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid. All sections are written and peer-reviewed by leading flow cytometry experts and immunologists, making this edition an essential and state-of-the-art handbook for basic and clinical researchers.
Collapse
Affiliation(s)
- Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Hyun-Dong Chang
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Institute for Biotechnology, Technische Universität, Berlin, Germany
| | - Andreas Radbruch
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Sergio Abrignani
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Richard Addo
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Immanuel Andrä
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Francesco Andreata
- Division of Immunology, Transplantation and Infectious Diseases, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eduardo Arranz
- Mucosal Immunology Lab, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - Petra Bacher
- Institute of Immunology, Christian-Albrechts Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Molecular Biology Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Sudipto Bari
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
- Center for Life Nano & Neuro Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
- Istituto Pasteur - Fondazione Cenci Bolognetti, Rome, Italy
| | | | - Dirk Baumjohann
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Cristian G. Beccaria
- Division of Immunology, Transplantation and Infectious Diseases, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - David Bernardo
- Mucosal Immunology Lab, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Dominic A. Boardman
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Jessica Borger
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - Chotima Böttcher
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leonie Brockmann
- Department of Microbiology & Immunology, Columbia University, New York City, USA
| | - Marie Burns
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Dirk H. Busch
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Garth Cameron
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Ilenia Cammarata
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Yinshui Chang
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Fernando Gabriel Chirdo
- Instituto de Estudios Inmunológicos y Fisiopatológicos - IIFP (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Eleni Christakou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Luka Čičin-Šain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Laura Cook
- BC Children’s Hospital Research Institute, Vancouver, Canada
- Department of Medicine, The University of British Columbia, Vancouver, Canada
| | - Alexandra J. Corbett
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca Cornelis
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Martin S. Davey
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Gabriele De Simone
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Michael Delacher
- Institute for Immunology, University Medical Center Mainz, Mainz, Germany
- Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Francesca Di Rosa
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - James Di Santo
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Inserm U1223, Paris, France
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Mucosal and Developmental Immunology, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Jun Dong
- Cell Biology, German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
| | - Thomas Dörner
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Department of Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Regine J. Dress
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Charles-Antoine Dutertre
- Institut National de la Sante Et de la Recherce Medicale (INSERM) U1015, Equipe Labellisee-Ligue Nationale contre le Cancer, Villejuif, France
| | - Sidonia B. G. Eckle
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Pascale Eede
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Maximilien Evrard
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | - Christine S. Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Markus Feuerer
- Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Simon Fillatreau
- Institut Necker Enfants Malades, INSERM U1151-CNRS, UMR8253, Paris, France
- Université de Paris, Paris Descartes, Faculté de Médecine, Paris, France
- AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - Aida Fiz-Lopez
- Mucosal Immunology Lab, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - Marie Follo
- Department of Medicine I, Lighthouse Core Facility, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gemma A. Foulds
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
- Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Julia Fröbel
- Immunology of Aging, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Nicola Gagliani
- Department of Medicine, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| | - Giovanni Galletti
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Anastasia Gangaev
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Natalio Garbi
- Institute of Molecular Medicine and Experimental Immunology, Faculty of Medicine, University of Bonn, Germany
| | - José Antonio Garrote
- Mucosal Immunology Lab, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
- Laboratory of Molecular Genetics, Servicio de Análisis Clínicos, Hospital Universitario Río Hortega, Gerencia Regional de Salud de Castilla y León (SACYL), Valladolid, Spain
| | - Jens Geginat
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Nicholas A. Gherardin
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Paola Gruarin
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
| | - Claudia Haftmann
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Leo Hansmann
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin (CVK), Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, Germany
| | - Christopher M. Harpur
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Adrian C. Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Guido Heine
- Division of Allergy, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Daniela Carolina Hernández
- Innate Immunity, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases, Rheumatology, Berlin, Germany
| | - Martin Herrmann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Oliver Hoelsken
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Mucosal and Developmental Immunology, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Qing Huang
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Samuel Huber
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johanna E. Huber
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Hundemer
- Department of Hematology, Oncology and Rheumatology, University Heidelberg, Heidelberg, Germany
| | - William Y. K. Hwang
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Department of Hematology, Singapore General Hospital, Singapore, Singapore
- Executive Offices, National Cancer Centre Singapore, Singapore
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCSS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sabine M. Ivison
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Peter K. Jani
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Baerbel Keller
- Department of Rheumatology and Clinical Immunology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nina Kessler
- Institute of Molecular Medicine and Experimental Immunology, Faculty of Medicine, University of Bonn, Germany
| | - Steven Ketelaars
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Laura Knop
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Jasmin Knopf
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Katja Kobow
- Department of Neuropathology, Universitätsklinikum Erlangen, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Heidelberg, Heidelberg, Germany
| | - H. Kristyanto
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andreas Krueger
- Institute for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jenny F. Kuehne
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Heike Kunze-Schumacher
- Institute for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | | | - Daniel Lenz
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Megan K. Levings
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, Canada
| | - Andreia C. Lino
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Heather M. Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Enrico Lugli
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Katherine N. MacDonald
- BC Children’s Hospital Research Institute, Vancouver, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, The University of British Columbia, Vancouver, Canada
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mala K. Maini
- Division of Infection & Immunity, Institute of Immunity & Transplantation, University College London, London, UK
| | - Florian Mair
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Calin Manta
- Department of Hematology, Oncology and Rheumatology, University Heidelberg, Heidelberg, Germany
| | - Rudolf Armin Manz
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Germany
| | | | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Henrik E. Mei
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Fritz Melchers
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Susanne Melzer
- Clinical Trial Center Leipzig, Leipzig University, Härtelstr.16, −18, Leipzig, 04107, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Leticia Monin
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Gabriele Multhoff
- Radiation Immuno-Oncology Group, Center for Translational Cancer Research (TranslaTUM), Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Luis Enrique Muñoz
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Miguel Muñoz-Ruiz
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Franziska Muscate
- Department of Medicine, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ambra Natalini
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
| | - Katrin Neumann
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lai Guan Ng
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Department of Microbiology & Immunology, Immunology Programme, Life Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Jana Niemz
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Samuele Notarbartolo
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
| | - Lennard Ostendorf
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Laura J. Pallett
- Division of Infection & Immunity, Institute of Immunity & Transplantation, University College London, London, UK
| | - Amit A. Patel
- Institut National de la Sante Et de la Recherce Medicale (INSERM) U1015, Equipe Labellisee-Ligue Nationale contre le Cancer, Villejuif, France
| | - Gulce Itir Percin
- Immunology of Aging, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Giovanna Peruzzi
- Center for Life Nano & Neuro Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - A. Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
- Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Katharina Pracht
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irma Pujol-Autonell
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
- Peter Gorer Department of Immunobiology, King’s College London, London, UK
| | - Nadia Pulvirenti
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
| | - Linda Quatrini
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Kylie M. Quinn
- School of Biomedical and Health Sciences, RMIT University, Bundorra, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Helena Radbruch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hefin Rhys
- Flow Cytometry Science Technology Platform, The Francis Crick Institute, London, UK
| | - Maria B. Rodrigo
- Institute of Molecular Medicine and Experimental Immunology, Faculty of Medicine, University of Bonn, Germany
| | - Chiara Romagnani
- Innate Immunity, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases, Rheumatology, Berlin, Germany
| | - Carina Saggau
- Institute of Immunology, Christian-Albrechts Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | | | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Lieke Sanderink
- Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christine Schauer
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Hans U. Scherer
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias Schiemann
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Frank A. Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kilian Schober
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Janina Schoen
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wolfgang Schuh
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Axel R. Schulz
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Sebastian Schulz
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Schulze
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Sonia Simonetti
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
| | - Jeeshan Singh
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katarzyna M. Sitnik
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Regina Stark
- Charité Universitätsmedizin Berlin – BIH Center for Regenerative Therapies, Berlin, Germany
- Sanquin Research – Adaptive Immunity, Amsterdam, The Netherlands
| | - Sarah Starossom
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christina Stehle
- Innate Immunity, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases, Rheumatology, Berlin, Germany
| | - Franziska Szelinski
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Department of Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Leonard Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Department of Microbiology & Immunology, Immunology Programme, Life Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Attila Tarnok
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
- Department of Precision Instrument, Tsinghua University, Beijing, China
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Julia Tornack
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Timothy I. M. Tree
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Jasper J. P. van Beek
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | - Chiara Vasco
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
| | - Nikita A. Verheyden
- Institute for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anouk von Borstel
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kirsten A. Ward-Hartstonge
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Waskow
- Immunology of Aging, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich-Schiller-University Jena, Jena, Germany
- Department of Medicine III, Technical University Dresden, Dresden, Germany
| | - Annika Wiedemann
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Department of Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anneke Wilharm
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - James Wing
- Immunology Frontier Research Center, Osaka University, Japan
| | - Oliver Wirz
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jens Wittner
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jennie H. M. Yang
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Juhao Yang
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
11
|
Human T-Cell Cloning by Limiting Dilution. Methods Mol Biol 2021. [PMID: 33928552 DOI: 10.1007/978-1-0716-1311-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Human T cells represent a heterogeneous population, including cells with different phenotypical and function properties. Despite, in the last years, several technologies were developed to investigate phenotypical properties of T cells at single cell level, in vitro T cell clone 's culture remains the only way to perform functional study on T cells at single cell levels. Here, we describe the method to obtain human T cell clones by limiting dilution in the presence of feeder cells and to maintain them in culture for further investigations.
Collapse
|
12
|
IL4I1 Is Expressed by Head-Neck Cancer-Derived Mesenchymal Stromal Cells and Contributes to Suppress T Cell Proliferation. J Clin Med 2021; 10:jcm10102111. [PMID: 34068423 PMCID: PMC8153554 DOI: 10.3390/jcm10102111] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/10/2021] [Accepted: 05/10/2021] [Indexed: 11/16/2022] Open
Abstract
Amino acids have a primary role in cancer metabolism. Beyond their primary biosynthetic role, they represent also an alternative fuel while their catabolites can influence the epigenetic control of gene expression and suppress anti-tumor immune responses. The accumulation of amino-acid derivatives in the tumor microenvironment depends not only on the activity of tumor cells, but also on stromal cells. In this study, we show that mesenchymal stromal cells derived from head–neck cancer express the amino acid oxidase IL4I1 that has been detected in different types of tumor cells. The catabolic products of IL4I1, H2O2, and kynurenines are known to suppress T cell response. We found that neutralization of IL4I1 activity can restore T cell proliferation. Thus, therapeutical strategies targeting enzymes involved in amino-acid catabolism may be helpful to contemporary block tumor cell migration and restore an efficacious anti-tumor immunity.
Collapse
|
13
|
Integrative computational approach identifies drug targets in CD4 + T-cell-mediated immune disorders. NPJ Syst Biol Appl 2021; 7:4. [PMID: 33483502 PMCID: PMC7822845 DOI: 10.1038/s41540-020-00165-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
CD4+ T cells provide adaptive immunity against pathogens and abnormal cells, and they are also associated with various immune-related diseases. CD4+ T cells’ metabolism is dysregulated in these pathologies and represents an opportunity for drug discovery and development. Genome-scale metabolic modeling offers an opportunity to accelerate drug discovery by providing high-quality information about possible target space in the context of a modeled disease. Here, we develop genome-scale models of naïve, Th1, Th2, and Th17 CD4+ T-cell subtypes to map metabolic perturbations in rheumatoid arthritis, multiple sclerosis, and primary biliary cholangitis. We subjected these models to in silico simulations for drug response analysis of existing FDA-approved drugs and compounds. Integration of disease-specific differentially expressed genes with altered reactions in response to metabolic perturbations identified 68 drug targets for the three autoimmune diseases. In vitro experimental validation, together with literature-based evidence, showed that modulation of fifty percent of identified drug targets suppressed CD4+ T cells, further increasing their potential impact as therapeutic interventions. Our approach can be generalized in the context of other diseases, and the metabolic models can be further used to dissect CD4+ T-cell metabolism.
Collapse
|
14
|
Uhlorn JA, Husband NA, Romero‐Aleshire MJ, Moffett C, Lindsey ML, Langlais PR, Brooks HL. CD4 + T Cell-Specific Proteomic Pathways Identified in Progression of Hypertension Across Postmenopausal Transition. J Am Heart Assoc 2021; 10:e018038. [PMID: 33410333 PMCID: PMC7955317 DOI: 10.1161/jaha.120.018038] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
Background Menopause is associated with an increase in the prevalence and severity of hypertension in women. Although premenopausal females are protected against T cell-dependent immune activation and development of angiotensin II (Ang II) hypertension, this protection is lost in postmenopausal females. Therefore, the current study hypothesized that specific CD4+ T cell pathways are regulated by sex hormones and Ang II to mediate progression from premenopausal protection to postmenopausal hypertension. Methods and Results Menopause was induced in C57BL/6 mice via repeated 4-vinylcyclohexene diepoxide injections, while premenopausal females received sesame oil vehicle. A subset of premenopausal mice and all menopausal mice were infused with Ang II for 14 days (Control, Ang II, Meno/Ang II). Proteomic and phosphoproteomic profiles of CD4+ T cells isolated from spleens were examined. Ang II markedly increased CD4+ T cell protein abundance and phosphorylation associated with DNA and histone methylation in both premenopausal and postmenopausal females. Compared with premenopausal T cells, Ang II infusion in menopausal mice increased T cell phosphorylation of MP2K2, an upstream regulator of ERK, and was associated with upregulated phosphorylation at ERK targeted sites. Additionally, Ang II infusion in menopausal mice decreased T cell phosphorylation of TLN1, a key regulator of IL-2Rα and FOXP3 expression. Conclusions These findings identify novel, distinct T cell pathways that influence T cell-mediated inflammation during postmenopausal hypertension.
Collapse
Affiliation(s)
- Joshua A. Uhlorn
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| | | | | | - Caitlin Moffett
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| | - Merry L. Lindsey
- Department of Cellular and Integrative PhysiologyCenter for Heart and Vascular ResearchNebraska‐Western Iowa Health Care SystemUniversity of Nebraska Medical Center and Research ServiceOmahaNE
| | - Paul R. Langlais
- Department of MedicineCollege of MedicineUniversity of ArizonaTucsonAZ
| | - Heddwen L. Brooks
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| |
Collapse
|
15
|
Monosomy 3 Influences Epithelial-Mesenchymal Transition Gene Expression in Uveal Melanoma Patients; Consequences for Liquid Biopsy. Int J Mol Sci 2020; 21:ijms21249651. [PMID: 33348918 PMCID: PMC7767066 DOI: 10.3390/ijms21249651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Despite outstanding advances in diagnosis and the treatment of primary uveal melanoma (UM), nearly 50% of UM patients develop metastases via hematogenous dissemination, driven by the epithelial-mesenchymal transition (EMT). Despite the failure in UM to date, a liquid biopsy may offer a feasible non-invasive approach for monitoring metastatic disease progression and addressing protracted dormancy. To detect circulating tumor cells (CTCs) in UM patients, we evaluated the mRNA expression of EMT-associated transcription factors in CD45-depleted blood fraction, using qRT-PCR. ddPCR was employed to assess UM-specific GNA11, GNAQ, PLCβ4, and CYSLTR2 mutations in plasma DNA. Moreover, microarray analysis was performed on total RNA isolated from tumor tissues to estimate the prognostic value of EMT-associated gene expression. In total, 42 primary UM and 11 metastatic patients were enrolled. All CD45-depleted samples were negative for CTC when compared to the peripheral blood fraction of 60 healthy controls. Tumor-specific mutations were detected in the plasma of 21.4% patients, merely, in 9.4% of primary UM, while 54.5% in metastatic patients. Unsupervised hierarchical clustering of differentially expressed EMT genes showed significant differences between monosomy 3 and disomy 3 tumors. Newly identified genes can serve as non-invasive prognostic biomarkers that can support therapeutic decisions.
Collapse
|
16
|
Kunkl M, Frascolla S, Amormino C, Volpe E, Tuosto L. T Helper Cells: The Modulators of Inflammation in Multiple Sclerosis. Cells 2020; 9:cells9020482. [PMID: 32093011 PMCID: PMC7072830 DOI: 10.3390/cells9020482] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by the progressive loss of axonal myelin in several areas of the central nervous system (CNS) that is responsible for clinical symptoms such as muscle spasms, optic neuritis, and paralysis. The progress made in more than one decade of research in animal models of MS for clarifying the pathophysiology of MS disease validated the concept that MS is an autoimmune inflammatory disorder caused by the recruitment in the CNS of self-reactive lymphocytes, mainly CD4+ T cells. Indeed, high levels of T helper (Th) cells and related cytokines and chemokines have been found in CNS lesions and in cerebrospinal fluid (CSF) of MS patients, thus contributing to the breakdown of the blood-brain barrier (BBB), the activation of resident astrocytes and microglia, and finally the outcome of neuroinflammation. To date, several types of Th cells have been discovered and designated according to the secreted lineage-defining cytokines. Interestingly, Th1, Th17, Th1-like Th17, Th9, and Th22 have been associated with MS. In this review, we discuss the role and interplay of different Th cell subpopulations and their lineage-defining cytokines in modulating the inflammatory responses in MS and the approved as well as the novel therapeutic approaches targeting T lymphocytes in the treatment of the disease.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Simone Frascolla
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Elisabetta Volpe
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| |
Collapse
|
17
|
Krummey SM, Hartigan CR, Liu D, Ford ML. CD28-Dependent CTLA-4 Expression Fine-Tunes the Activation of Human Th17 Cells. iScience 2020; 23:100912. [PMID: 32203908 PMCID: PMC7096747 DOI: 10.1016/j.isci.2020.100912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 09/26/2019] [Accepted: 02/10/2020] [Indexed: 01/05/2023] Open
Abstract
Previous work has demonstrated that Th17 memory cells but not Th1 cells are resistant to CD28/CTLA-4 blockade with CTLA-4 Ig, leading us to investigate the individual roles of the CD28 and CTLA-4 cosignaling pathways on Th1 versus Th17 cells. We found that selective CD28 blockade with a domain antibody (dAb) inhibited Th1 cells but surprisingly augmented Th17 responses. CD28 agonism resulted in a profound increase in CTLA-4 expression in Th17 cells as compared with Th1 cells. Consistent with these findings, inhibition of the CD28 signaling protein AKT revealed that CTLA-4 expression on Th17 cells was more significantly reduced by AKT inhibition relative to CTLA-4 expression on Th17 cells. Finally, we found that FOXO1 and FOXO3 overexpression restrained high expression of CTLA-4 on Th17 cells but not Th1 cells. This study demonstrates that the heterogeneity of the CD4+ T cell compartment has implications for the immunomodulation of pathologic T cell responses. CD28 blockade resulted in augmentation of human Th17 cells relative to Th1 cells Th17 polarized mice exhibited graft rejection in the presence of CD28 blockade A significant portion of Th17 cell CTLA-4 expression was induced by CD28 ligation Overexpression of FOXO1 or FOXO3 inhibited Th17 cell CTLA-4 expression
Collapse
Affiliation(s)
- Scott M Krummey
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Christina R Hartigan
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Danya Liu
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Mandy L Ford
- Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
18
|
Mazzoni A, Maggi L, Liotta F, Cosmi L, Annunziato F. Biological and clinical significance of T helper 17 cell plasticity. Immunology 2019; 158:287-295. [PMID: 31566706 DOI: 10.1111/imm.13124] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/16/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022] Open
Abstract
Mature T helper (Th) effector cells originate following antigen recognition by naive T precursors. The maturation process is accompanied by the acquisition of specific effector functions that distinguish at least three different T helper subsets: Th1, Th2 and Th17. In general, maturation of somatic cells is accompanied by terminal differentiation. However, accumulating evidence shows that effector T cells retain a certain degree of plasticity. This is especially true for Th17 cells, which have been shown to converge towards other phenotypes in response to specific microenvironmental pressure. In this review we will discuss the experimental evidence that supports the hypothesis of Th17 plasticity, with particular emphasis on the generation of Th17-derived 'non-classic' Th1 cells, and the molecular networks that control it. Moreover, we will consider why Th17 plasticity is important for host protection, but also why it can have pathogenic functions during chronic inflammation. Regarding the last point, we will discuss a possible role for biological drugs in the control of Th17 plasticity and disease course.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
19
|
Cossarizza A, Chang HD, Radbruch A, Acs A, Adam D, Adam-Klages S, Agace WW, Aghaeepour N, Akdis M, Allez M, Almeida LN, Alvisi G, Anderson G, Andrä I, Annunziato F, Anselmo A, Bacher P, Baldari CT, Bari S, Barnaba V, Barros-Martins J, Battistini L, Bauer W, Baumgart S, Baumgarth N, Baumjohann D, Baying B, Bebawy M, Becher B, Beisker W, Benes V, Beyaert R, Blanco A, Boardman DA, Bogdan C, Borger JG, Borsellino G, Boulais PE, Bradford JA, Brenner D, Brinkman RR, Brooks AES, Busch DH, Büscher M, Bushnell TP, Calzetti F, Cameron G, Cammarata I, Cao X, Cardell SL, Casola S, Cassatella MA, Cavani A, Celada A, Chatenoud L, Chattopadhyay PK, Chow S, Christakou E, Čičin-Šain L, Clerici M, Colombo FS, Cook L, Cooke A, Cooper AM, Corbett AJ, Cosma A, Cosmi L, Coulie PG, Cumano A, Cvetkovic L, Dang VD, Dang-Heine C, Davey MS, Davies D, De Biasi S, Del Zotto G, Cruz GVD, Delacher M, Bella SD, Dellabona P, Deniz G, Dessing M, Di Santo JP, Diefenbach A, Dieli F, Dolf A, Dörner T, Dress RJ, Dudziak D, Dustin M, Dutertre CA, Ebner F, Eckle SBG, Edinger M, Eede P, Ehrhardt GR, Eich M, Engel P, Engelhardt B, Erdei A, Esser C, Everts B, Evrard M, Falk CS, Fehniger TA, Felipo-Benavent M, Ferry H, Feuerer M, Filby A, Filkor K, Fillatreau S, Follo M, Förster I, Foster J, Foulds GA, Frehse B, Frenette PS, Frischbutter S, Fritzsche W, Galbraith DW, Gangaev A, Garbi N, Gaudilliere B, Gazzinelli RT, Geginat J, Gerner W, Gherardin NA, Ghoreschi K, Gibellini L, Ginhoux F, Goda K, Godfrey DI, Goettlinger C, González-Navajas JM, Goodyear CS, Gori A, Grogan JL, Grummitt D, Grützkau A, Haftmann C, Hahn J, Hammad H, Hämmerling G, Hansmann L, Hansson G, Harpur CM, Hartmann S, Hauser A, Hauser AE, Haviland DL, Hedley D, Hernández DC, Herrera G, Herrmann M, Hess C, Höfer T, Hoffmann P, Hogquist K, Holland T, Höllt T, Holmdahl R, Hombrink P, Houston JP, Hoyer BF, Huang B, Huang FP, Huber JE, Huehn J, Hundemer M, Hunter CA, Hwang WYK, Iannone A, Ingelfinger F, Ivison SM, Jäck HM, Jani PK, Jávega B, Jonjic S, Kaiser T, Kalina T, Kamradt T, Kaufmann SHE, Keller B, Ketelaars SLC, Khalilnezhad A, Khan S, Kisielow J, Klenerman P, Knopf J, Koay HF, Kobow K, Kolls JK, Kong WT, Kopf M, Korn T, Kriegsmann K, Kristyanto H, Kroneis T, Krueger A, Kühne J, Kukat C, Kunkel D, Kunze-Schumacher H, Kurosaki T, Kurts C, Kvistborg P, Kwok I, Landry J, Lantz O, Lanuti P, LaRosa F, Lehuen A, LeibundGut-Landmann S, Leipold MD, Leung LY, Levings MK, Lino AC, Liotta F, Litwin V, Liu Y, Ljunggren HG, Lohoff M, Lombardi G, Lopez L, López-Botet M, Lovett-Racke AE, Lubberts E, Luche H, Ludewig B, Lugli E, Lunemann S, Maecker HT, Maggi L, Maguire O, Mair F, Mair KH, Mantovani A, Manz RA, Marshall AJ, Martínez-Romero A, Martrus G, Marventano I, Maslinski W, Matarese G, Mattioli AV, Maueröder C, Mazzoni A, McCluskey J, McGrath M, McGuire HM, McInnes IB, Mei HE, Melchers F, Melzer S, Mielenz D, Miller SD, Mills KH, Minderman H, Mjösberg J, Moore J, Moran B, Moretta L, Mosmann TR, Müller S, Multhoff G, Muñoz LE, Münz C, Nakayama T, Nasi M, Neumann K, Ng LG, Niedobitek A, Nourshargh S, Núñez G, O’Connor JE, Ochel A, Oja A, Ordonez D, Orfao A, Orlowski-Oliver E, Ouyang W, Oxenius A, Palankar R, Panse I, Pattanapanyasat K, Paulsen M, Pavlinic D, Penter L, Peterson P, Peth C, Petriz J, Piancone F, Pickl WF, Piconese S, Pinti M, Pockley AG, Podolska MJ, Poon Z, Pracht K, Prinz I, Pucillo CEM, Quataert SA, Quatrini L, Quinn KM, Radbruch H, Radstake TRDJ, Rahmig S, Rahn HP, Rajwa B, Ravichandran G, Raz Y, Rebhahn JA, Recktenwald D, Reimer D, e Sousa CR, Remmerswaal EB, Richter L, Rico LG, Riddell A, Rieger AM, Robinson JP, Romagnani C, Rubartelli A, Ruland J, Saalmüller A, Saeys Y, Saito T, Sakaguchi S, de-Oyanguren FS, Samstag Y, Sanderson S, Sandrock I, Santoni A, Sanz RB, Saresella M, Sautes-Fridman C, Sawitzki B, Schadt L, Scheffold A, Scherer HU, Schiemann M, Schildberg FA, Schimisky E, Schlitzer A, Schlosser J, Schmid S, Schmitt S, Schober K, Schraivogel D, Schuh W, Schüler T, Schulte R, Schulz AR, Schulz SR, Scottá C, Scott-Algara D, Sester DP, Shankey TV, Silva-Santos B, Simon AK, Sitnik KM, Sozzani S, Speiser DE, Spidlen J, Stahlberg A, Stall AM, Stanley N, Stark R, Stehle C, Steinmetz T, Stockinger H, Takahama Y, Takeda K, Tan L, Tárnok A, Tiegs G, Toldi G, Tornack J, Traggiai E, Trebak M, Tree TI, Trotter J, Trowsdale J, Tsoumakidou M, Ulrich H, Urbanczyk S, van de Veen W, van den Broek M, van der Pol E, Van Gassen S, Van Isterdael G, van Lier RA, Veldhoen M, Vento-Asturias S, Vieira P, Voehringer D, Volk HD, von Borstel A, von Volkmann K, Waisman A, Walker RV, Wallace PK, Wang SA, Wang XM, Ward MD, Ward-Hartstonge KA, Warnatz K, Warnes G, Warth S, Waskow C, Watson JV, Watzl C, Wegener L, Weisenburger T, Wiedemann A, Wienands J, Wilharm A, Wilkinson RJ, Willimsky G, Wing JB, Winkelmann R, Winkler TH, Wirz OF, Wong A, Wurst P, Yang JHM, Yang J, Yazdanbakhsh M, Yu L, Yue A, Zhang H, Zhao Y, Ziegler SM, Zielinski C, Zimmermann J, Zychlinsky A. Guidelines for the use of flow cytometry and cell sorting in immunological studies (second edition). Eur J Immunol 2019; 49:1457-1973. [PMID: 31633216 PMCID: PMC7350392 DOI: 10.1002/eji.201970107] [Citation(s) in RCA: 710] [Impact Index Per Article: 142.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
These guidelines are a consensus work of a considerable number of members of the immunology and flow cytometry community. They provide the theory and key practical aspects of flow cytometry enabling immunologists to avoid the common errors that often undermine immunological data. Notably, there are comprehensive sections of all major immune cell types with helpful Tables detailing phenotypes in murine and human cells. The latest flow cytometry techniques and applications are also described, featuring examples of the data that can be generated and, importantly, how the data can be analysed. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid, all written and peer-reviewed by leading experts in the field, making this an essential research companion.
Collapse
Affiliation(s)
- Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, Univ. of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Hyun-Dong Chang
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Andreas Acs
- Department of Biology, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Sabine Adam-Klages
- Institut für Transfusionsmedizin, Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - William W. Agace
- Mucosal Immunology group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
- Immunology Section, Lund University, Lund, Sweden
| | - Nima Aghaeepour
- Departments of Anesthesiology, Pain and Perioperative Medicine; Biomedical Data Sciences; and Pediatrics, Stanford University, Stanford, CA, USA
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Matthieu Allez
- Université de Paris, Institut de Recherche Saint-Louis, INSERM U1160, and Gastroenterology Department, Hôpital Saint-Louis – APHP, Paris, France
| | | | - Giorgia Alvisi
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Italy
| | | | - Immanuel Andrä
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Achille Anselmo
- Flow Cytometry Core, Humanitas Clinical and Research Center, Milan, Italy
| | - Petra Bacher
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
- Institut für Klinische Molekularbiologie, Christian-Albrechts Universität zu Kiel, Germany
| | | | - Sudipto Bari
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
- Istituto Pasteur - Fondazione Cenci Bolognetti, Rome, Italy
| | | | | | - Wolfgang Bauer
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sabine Baumgart
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Nicole Baumgarth
- Center for Comparative Medicine & Dept. Pathology, Microbiology & Immunology, University of California, Davis, CA, USA
| | - Dirk Baumjohann
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Planegg-Martinsried, Germany
| | - Bianka Baying
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Mary Bebawy
- Discipline of Pharmacy, Graduate School of Health, The University of Technology Sydney, Sydney, NSW, Australia
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Switzerland
| | - Wolfgang Beisker
- Flow Cytometry Laboratory, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health, München, Germany
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Rudi Beyaert
- Department of Biomedical Molecular Biology, Center for Inflammation Research, Ghent University - VIB, Ghent, Belgium
| | - Alfonso Blanco
- Flow Cytometry Core Technologies, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Dominic A. Boardman
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Erlangen, Germany
- Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Medical Immunology Campus Erlangen, Erlangen, Germany
| | - Jessica G. Borger
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - Giovanna Borsellino
- Neuroimmunology and Flow Cytometry Units, Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Philip E. Boulais
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Bronx, New York, USA
| | | | - Dirk Brenner
- Luxembourg Institute of Health, Department of Infection and Immunity, Experimental and Molecular Immunology, Esch-sur-Alzette, Luxembourg
- Odense University Hospital, Odense Research Center for Anaphylaxis, University of Southern Denmark, Department of Dermatology and Allergy Center, Odense, Denmark
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Ryan R. Brinkman
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
| | - Anna E. S. Brooks
- University of Auckland, School of Biological Sciences, Maurice Wilkins Center, Auckland, New Zealand
| | - Dirk H. Busch
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
- Focus Group “Clinical Cell Processing and Purification”, Institute for Advanced Study, Technische Universität München, Munich, Germany
| | - Martin Büscher
- Biophysics, R&D Engineering, Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Timothy P. Bushnell
- Department of Pediatrics and Shared Resource Laboratories, University of Rochester Medical Center, Rochester, NY, USA
| | - Federica Calzetti
- University of Verona, Department of Medicine, Section of General Pathology, Verona, Italy
| | - Garth Cameron
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Ilenia Cammarata
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology, Nankai University, Tianjin, China
| | - Susanna L. Cardell
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Stefano Casola
- The FIRC Institute of Molecular Oncology (FOM), Milan, Italy
| | - Marco A. Cassatella
- University of Verona, Department of Medicine, Section of General Pathology, Verona, Italy
| | - Andrea Cavani
- National Institute for Health, Migration and Poverty (INMP), Rome, Italy
| | - Antonio Celada
- Macrophage Biology Group, School of Biology, University of Barcelona, Barcelona, Spain
| | - Lucienne Chatenoud
- Université Paris Descartes, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | | | - Sue Chow
- Divsion of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Eleni Christakou
- Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institutes of Health Research Biomedical Research Centre at Guy’s and St. Thomas’ National Health Service, Foundation Trust and King’s College London, UK
| | - Luka Čičin-Šain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Department of Physiopathology and Transplants, University of Milan, Milan, Italy
- Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | | | - Laura Cook
- BC Children’s Hospital Research Institute, Vancouver, Canada
- Department of Medicine, The University of British Columbia, Vancouver, Canada
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Andrea M. Cooper
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Alexandra J. Corbett
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Antonio Cosma
- National Cytometry Platform, Luxembourg Institute of Health, Department of Infection and Immunity, Esch-sur-Alzette, Luxembourg
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pierre G. Coulie
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Ana Cumano
- Unit Lymphopoiesis, Department of Immunology, Institut Pasteur, Paris, France
| | - Ljiljana Cvetkovic
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Van Duc Dang
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Chantip Dang-Heine
- Clinical Research Unit, Berlin Institute of Health (BIH), Charite Universitätsmedizin Berlin, Berlin, Germany
| | - Martin S. Davey
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | - Derek Davies
- Flow Cytometry Scientific Technology Platform, The Francis Crick Institute, London, UK
| | - Sara De Biasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, Univ. of Modena and Reggio Emilia, Modena, Italy
| | | | - Gelo Victoriano Dela Cruz
- Novo Nordisk Foundation Center for Stem Cell Biology – DanStem, University of Copenhagen, Copenhagen, Denmark
| | - Michael Delacher
- Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
- Chair for Immunology, University Regensburg, Germany
| | - Silvia Della Bella
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Paolo Dellabona
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Günnur Deniz
- Istanbul University, Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul, Turkey
| | | | - James P. Di Santo
- Innate Immunty Unit, Department of Immunology, Institut Pasteur, Paris, France
- Institut Pasteur, Inserm U1223, Paris, France
| | - Andreas Diefenbach
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Francesco Dieli
- University of Palermo, Central Laboratory of Advanced Diagnosis and Biomedical Research, Department of Biomedicine, Neurosciences and Advanced Diagnostics, Palermo, Italy
| | - Andreas Dolf
- Flow Cytometry Core Facility, Institute of Experimental Immunology, University of Bonn, Bonn, Germany
| | - Thomas Dörner
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Dept. Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Germany
| | - Regine J. Dress
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Michael Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Charles-Antoine Dutertre
- Program in Emerging Infectious Disease, Duke-NUS Medical School, Singapore
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
| | - Friederike Ebner
- Institute of Immunology, Centre for Infection Medicine, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Sidonia B. G. Eckle
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Matthias Edinger
- Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Germany
| | - Pascale Eede
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Germany
| | | | - Marcus Eich
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Pablo Engel
- University of Barcelona, Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Barcelona, Spain
| | | | - Anna Erdei
- Department of Immunology, University L. Eotvos, Budapest, Hungary
| | - Charlotte Esser
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maximilien Evrard
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
| | - Christine S. Falk
- Institute of Transplant Immunology, Hannover Medical School, MHH, Hannover, Germany
| | - Todd A. Fehniger
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mar Felipo-Benavent
- Laboratory of Cytomics, Joint Research Unit CIPF-UVEG, Principe Felipe Research Center, Valencia, Spain
| | - Helen Ferry
- Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Markus Feuerer
- Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
- Chair for Immunology, University Regensburg, Germany
| | - Andrew Filby
- The Flow Cytometry Core Facility, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | | | - Simon Fillatreau
- Institut Necker-Enfants Malades, Université Paris Descartes Sorbonne Paris Cité, Faculté de Médecine, AP-HP, Hôpital Necker Enfants Malades, INSERM U1151-CNRS UMR 8253, Paris, France
| | - Marie Follo
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Universitaetsklinikum FreiburgLighthouse Core Facility, Zentrum für Translationale Zellforschung, Klinik für Innere Medizin I, Freiburg, Germany
| | - Irmgard Förster
- Immunology and Environment, LIMES Institute, University of Bonn, Bonn, Germany
| | | | - Gemma A. Foulds
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Britta Frehse
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Germany
| | - Paul S. Frenette
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Stefan Frischbutter
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Dermatology, Venereology and Allergology
| | - Wolfgang Fritzsche
- Nanobiophotonics Department, Leibniz Institute of Photonic Technology (IPHT), Jena, Germany
| | - David W. Galbraith
- School of Plant Sciences and Bio5 Institute, University of Arizona, Tucson, USA
- Honorary Dean of Life Sciences, Henan University, Kaifeng, China
| | - Anastasia Gangaev
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Natalio Garbi
- Institute of Experimental Immunology, University of Bonn, Germany
| | - Brice Gaudilliere
- Stanford Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, CA, USA
| | - Ricardo T. Gazzinelli
- Fundação Oswaldo Cruz - Minas, Laboratory of Immunopatology, Belo Horizonte, MG, Brazil
- Department of Mecicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jens Geginat
- INGM - Fondazione Istituto Nazionale di Genetica Molecolare “Ronmeo ed Enrica Invernizzi”, Milan, Italy
| | - Wilhelm Gerner
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Austria
- Christian Doppler Laboratory for Optimized Prediction of Vaccination Success in Pigs, Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Austria
| | - Nicholas A. Gherardin
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lara Gibellini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, Univ. of Modena and Reggio Emilia, Modena, Italy
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keisuke Goda
- Department of Bioengineering, University of California, Los Angeles, California, USA
- Department of Chemistry, University of Tokyo, Tokyo, Japan
- Institute of Technological Sciences, Wuhan University, Wuhan, China
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | | | - Jose M. González-Navajas
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd), Madrid, Spain
| | - Carl S. Goodyear
- Institute of Infection Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow Biomedical Research Centre, Glasgow, UK
| | - Andrea Gori
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, University of Milan
| | - Jane L. Grogan
- Cancer Immunology Research, Genentech, South San Francisco, CA, USA
| | | | - Andreas Grützkau
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Claudia Haftmann
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jonas Hahn
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen
| | - Hamida Hammad
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Zwijnaarde, Belgium
| | | | - Leo Hansmann
- Berlin Institute of Health (BIH), Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, Berlin, Germany
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Goran Hansson
- Department of Medicine and Center for Molecular Medicine at Karolinska University Hospital, Solna, Sweden
| | | | - Susanne Hartmann
- Institute of Immunology, Centre for Infection Medicine, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Andrea Hauser
- Department of Internal Medicine III, University Hospital Regensburg, Germany
| | - Anja E. Hauser
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin
- Department of Rheumatology and Clinical Immunology, Berlin Institute of Health, Berlin, Germany
| | - David L. Haviland
- Flow Cytometry, Houston Methodist Hospital Research Institute, Houston, TX, USA
| | - David Hedley
- Divsion of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Daniela C. Hernández
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Medical Department I, Division of Gastroenterology, Infectiology and Rheumatology, Berlin, Germany
| | - Guadalupe Herrera
- Cytometry Service, Incliva Foundation. Clinic Hospital and Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Martin Herrmann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen
| | - Christoph Hess
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Thomas Höfer
- German Cancer Research Center (DKFZ), Division of Theoretical Systems Biology, Heidelberg, Germany
| | - Petra Hoffmann
- Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Germany
| | - Kristin Hogquist
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Tristan Holland
- Institute of Experimental Immunology, University of Bonn, Germany
| | - Thomas Höllt
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, The Netherlands
- Computer Graphics and Visualization, Department of Intelligent Systems, TU Delft, Delft, The Netherlands
| | | | - Pleun Hombrink
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jessica P. Houston
- Department of Chemical & Materials Engineering, New Mexico State University, Las Cruces, NM, USA
| | - Bimba F. Hoyer
- Rheumatologie/Klinische Immunologie, Klinik für Innere Medizin I und Exzellenzzentrum Entzündungsmedizin, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Bo Huang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, China
| | - Fang-Ping Huang
- Institute for Advanced Study (IAS), Shenzhen University, Shenzhen, China
| | - Johanna E. Huber
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Planegg-Martinsried, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Hundemer
- Department of Hematology, Oncology and Rheumatology, University Heidelberg, Heidelberg, Germany
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William Y. K. Hwang
- Department of Hematology, Singapore General Hospital, Singapore
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore
- Executive Offices, National Cancer Centre Singapore, Singapore
| | - Anna Iannone
- Department of Diagnostic Medicine, Clinical and Public Health, Univ. of Modena and Reggio Emilia, Modena, Italy
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sabine M Ivison
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Peter K. Jani
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Beatriz Jávega
- Laboratory of Cytomics, Joint Research Unit CIPF-UVEG, Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain
| | - Stipan Jonjic
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Toralf Kaiser
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Tomas Kalina
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Thomas Kamradt
- Jena University Hospital, Institute of Immunology, Jena, Germany
| | | | - Baerbel Keller
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Steven L. C. Ketelaars
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ahad Khalilnezhad
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Srijit Khan
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Jan Kisielow
- Institute of Molecular Health Sciences, ETH Zurich, Zürich, Switzerland
| | - Paul Klenerman
- Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jasmin Knopf
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Katja Kobow
- Department of Neuropathology, Universitätsklinikum Erlangen, Germany
| | - Jay K. Kolls
- John W Deming Endowed Chair in Internal Medicine, Center for Translational Research in Infection and Inflammation Tulane School of Medicine, New Orleans, LA, USA
| | - Wan Ting Kong
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zurich, Zürich, Switzerland
| | - Thomas Korn
- Department of Neurology, Technical University of Munich, Munich, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Heidelberg, Heidelberg, Germany
| | - Hendy Kristyanto
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas Kroneis
- Division of Cell Biology, Histology & Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Andreas Krueger
- Institute for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jenny Kühne
- Institute of Transplant Immunology, Hannover Medical School, MHH, Hannover, Germany
| | - Christian Kukat
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Désirée Kunkel
- Flow & Mass Cytometry Core Facility, Charité - Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany
- BCRT Flow Cytometry Lab, Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin
| | - Heike Kunze-Schumacher
- Institute for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Tomohiro Kurosaki
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Christian Kurts
- Institute of Experimental Immunology, University of Bonn, Germany
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Jonathan Landry
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Olivier Lantz
- INSERM U932, PSL University, Institut Curie, Paris, France
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Francesca LaRosa
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Agnès Lehuen
- Institut Cochin, CNRS8104, INSERM1016, Department of Endocrinology, Metabolism and Diabetes, Université de Paris, Paris, France
| | | | - Michael D. Leipold
- The Human Immune Monitoring Center (HIMC), Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, CA, USA
| | - Leslie Y.T. Leung
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Megan K. Levings
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, Canada
| | - Andreia C. Lino
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Dept. Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Germany
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Yanling Liu
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, ANA Futura, Karolinska Institutet, Stockholm, Sweden
| | - Michael Lohoff
- Inst. f. Med. Mikrobiology and Hospital Hygiene, University of Marburg, Germany
| | - Giovanna Lombardi
- King’s College London, “Peter Gorer” Department of Immunobiology, London, UK
| | | | - Miguel López-Botet
- IMIM(Hospital de Mar Medical Research Institute), University Pompeu Fabra, Barcelona, Spain
| | - Amy E. Lovett-Racke
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Erik Lubberts
- Department of Rheumatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Herve Luche
- Centre d’Immunophénomique - CIPHE (PHENOMIN), Aix Marseille Université (UMS3367), Inserm (US012), CNRS (UMS3367), Marseille, France
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Italy
- Flow Cytometry Core, Humanitas Clinical and Research Center, Milan, Italy
| | - Sebastian Lunemann
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Holden T. Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Orla Maguire
- Flow and Image Cytometry Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Florian Mair
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
| | - Kerstin H. Mair
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Austria
- Christian Doppler Laboratory for Optimized Prediction of Vaccination Success in Pigs, Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Austria
| | - Alberto Mantovani
- Istituto Clinico Humanitas IRCCS and Humanitas University, Pieve Emanuele, Milan, Italy
- William Harvey Research Institute, Queen Mary University, London, United Kingdom
| | - Rudolf A. Manz
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Germany
| | - Aaron J. Marshall
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | | | - Glòria Martrus
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Ivana Marventano
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Wlodzimierz Maslinski
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Department of Pathophysiology and Immunology, Warsaw, Poland
| | - Giuseppe Matarese
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecologie Mediche, Università di Napoli Federico II and Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - Anna Vittoria Mattioli
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, Univ. of Modena and Reggio Emilia, Modena, Italy
- Lab of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Christian Maueröder
- Cell Clearance in Health and Disease Lab, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Mairi McGrath
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Helen M. McGuire
- Ramaciotti Facility for Human Systems Biology, and Discipline of Pathology, The University of Sydney, Camperdown, Australia
| | - Iain B. McInnes
- Institute of Infection Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow Biomedical Research Centre, Glasgow, UK
| | - Henrik E. Mei
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Fritz Melchers
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Susanne Melzer
- Clinical Trial Center Leipzig, University Leipzig, Leipzig, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stephen D. Miller
- Interdepartmental Immunobiology Center, Dept. of Microbiology-Immunology, Northwestern Univ. Medical School, Chicago, IL, USA
| | - Kingston H.G. Mills
- Trinity College Dublin, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Hans Minderman
- Flow and Image Cytometry Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine Huddinge, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical and Experimental Medine, Linköping University, Linköping, Sweden
| | - Jonni Moore
- Abramson Cancer Center Flow Cytometry and Cell Sorting Shared Resource, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Barry Moran
- Trinity College Dublin, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesu Children’s Hospital, Rome, Italy
| | - Tim R. Mosmann
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Susann Müller
- Centre for Environmental Research - UFZ, Department Environmental Microbiology, Leipzig, Germany
| | - Gabriele Multhoff
- Institute for Innovative Radiotherapy (iRT), Experimental Immune Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Radiation Immuno-Oncology Group, Center for Translational Cancer Research Technische Universität München (TranslaTUM), Klinikum rechts der Isar, Munich, Germany
| | - Luis Enrique Muñoz
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen
| | - Christian Münz
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Switzerland
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba city, Chiba, Japan
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, Univ. of Modena and Reggio Emilia, Modena, Italy
| | - Katrin Neumann
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
- Discipline of Dermatology, University of Sydney, Sydney, New South Wales, Australia
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Antonia Niedobitek
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Sussan Nourshargh
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, the University of Michigan, Ann Arbor, Michigan, USA
| | - José-Enrique O’Connor
- Laboratory of Cytomics, Joint Research Unit CIPF-UVEG, Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain
| | - Aaron Ochel
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Oja
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Diana Ordonez
- Flow Cytometry Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Alberto Orfao
- Department of Medicine, Cancer Research Centre (IBMCC-CSIC/USAL), Cytometry Service, University of Salamanca, CIBERONC and Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Eva Orlowski-Oliver
- Burnet Institute, AMREP Flow Cytometry Core Facility, Melbourne, Victoria, Australia
| | - Wenjun Ouyang
- Inflammation and Oncology, Research, Amgen Inc, South San Francisco, USA
| | | | - Raghavendra Palankar
- Department of Transfusion Medicine, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Isabel Panse
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Kovit Pattanapanyasat
- Center of Excellence for Flow Cytometry, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Malte Paulsen
- Flow Cytometry Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Dinko Pavlinic
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Livius Penter
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Christian Peth
- Biophysics, R&D Engineering, Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Jordi Petriz
- Functional Cytomics Group, Josep Carreras Leukaemia Research Institute, Campus ICO-Germans Trias i Pujol, Universitat Autònoma de Barcelona, UAB, Badalona, Spain
| | - Federica Piancone
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Silvia Piconese
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
- Istituto Pasteur - Fondazione Cenci Bolognetti, Rome, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - A. Graham Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
- Chromocyte Limited, Electric Works, Sheffield, UK
| | - Malgorzata Justyna Podolska
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen
- Department for Internal Medicine 3, Institute for Rheumatology and Immunology, AG Munoz, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Zhiyong Poon
- Department of Hematology, Singapore General Hospital, Singapore
| | - Katharina Pracht
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Sally A. Quataert
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Linda Quatrini
- Department of Immunology, IRCCS Bambino Gesu Children’s Hospital, Rome, Italy
| | - Kylie M. Quinn
- School of Biomedical and Health Sciences, RMIT University, Bundoora, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Helena Radbruch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Germany
| | - Tim R. D. J. Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Susann Rahmig
- Regeneration in Hematopoiesis, Leibniz-Institute on Aging, Fritz-Lipmann-Institute (FLI), Jena, Germany
| | - Hans-Peter Rahn
- Preparative Flow Cytometry, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Bartek Rajwa
- Bindley Biosciences Center, Purdue University, West Lafayette, IN, USA
| | - Gevitha Ravichandran
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yotam Raz
- Department of Internal Medicine, Groene Hart Hospital, Gouda, The Netherlands
| | - Jonathan A. Rebhahn
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Dorothea Reimer
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | - Ester B.M. Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lisa Richter
- Core Facility Flow Cytometry, Biomedical Center, Ludwig-Maximilians-University Munich, Germany
| | - Laura G. Rico
- Functional Cytomics Group, Josep Carreras Leukaemia Research Institute, Campus ICO-Germans Trias i Pujol, Universitat Autònoma de Barcelona, UAB, Badalona, Spain
| | - Andy Riddell
- Flow Cytometry Scientific Technology Platform, The Francis Crick Institute, London, UK
| | - Aja M. Rieger
- Department of Medical Microbiology and Immunology, University of Alberta, Alberta, Canada
| | - J. Paul Robinson
- Purdue University Cytometry Laboratories, Purdue University, West Lafayette, IN, USA
| | - Chiara Romagnani
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Medical Department I, Division of Gastroenterology, Infectiology and Rheumatology, Berlin, Germany
| | - Anna Rubartelli
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Fakultät für Medizin, Technische Universität München, München, Germany
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Austria
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Takashi Saito
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shimon Sakaguchi
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Francisco Sala de-Oyanguren
- Flow Cytometry Facility, Ludwig Cancer Institute, Faculty of Medicine and Biology, University of Lausanne, Epalinges, Switzerland
| | - Yvonne Samstag
- Heidelberg University, Institute of Immunology, Section of Molecular Immunology, Heidelberg, Germany
| | - Sharon Sanderson
- Translational Immunology Laboratory, NIHR BRC, University of Oxford, Kennedy Institute of Rheumatology, Oxford, UK
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, IRCCS, Neuromed, Pozzilli, Italy
| | - Ramon Bellmàs Sanz
- Institute of Transplant Immunology, Hannover Medical School, MHH, Hannover, Germany
| | - Marina Saresella
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | | | - Birgit Sawitzki
- Charité – Universitätsmedizin Berlin, and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Germany
| | - Linda Schadt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Switzerland
| | - Alexander Scheffold
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Hans U. Scherer
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias Schiemann
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Frank A. Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Andreas Schlitzer
- Quantitative Systems Biology, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Josephine Schlosser
- Institute of Immunology, Centre for Infection Medicine, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Stephan Schmid
- Internal Medicine I, University Hospital Regensburg, Germany
| | - Steffen Schmitt
- Flow Cytometry Core Facility, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Kilian Schober
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Daniel Schraivogel
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Wolfgang Schuh
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Reiner Schulte
- University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Axel Ronald Schulz
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Sebastian R. Schulz
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Cristiano Scottá
- King’s College London, “Peter Gorer” Department of Immunobiology, London, UK
| | - Daniel Scott-Algara
- Institut Pasteur, Cellular Lymphocytes Biology, Immunology Departement, Paris, France
| | - David P. Sester
- TRI Flow Cytometry Suite (TRI.fcs), Translational Research Institute, Wooloongabba, QLD, Australia
| | | | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | | | - Katarzyna M. Sitnik
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Silvano Sozzani
- Dept. Molecular Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniel E. Speiser
- Department of Oncology, University of Lausanne and CHUV, Epalinges, Switzerland
| | | | - Anders Stahlberg
- Lundberg Laboratory for Cancer, Department of Pathology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | | | - Natalie Stanley
- Departments of Anesthesiology, Pain and Perioperative Medicine; Biomedical Data Sciences; and Pediatrics, Stanford University, Stanford, CA, USA
| | - Regina Stark
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Christina Stehle
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Medical Department I, Division of Gastroenterology, Infectiology and Rheumatology, Berlin, Germany
| | - Tobit Steinmetz
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Kiyoshi Takeda
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Leonard Tan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Attila Tárnok
- Departement for Therapy Validation, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
- Department of Precision Instruments, Tsinghua University, Beijing, China
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Julia Tornack
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- BioGenes GmbH, Berlin, Germany
| | - Elisabetta Traggiai
- Novartis Biologics Center, Mechanistic Immunology Unit, Novartis Institute for Biomedical Research, NIBR, Basel, Switzerland
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, PA, United States
| | - Timothy I.M. Tree
- Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institutes of Health Research Biomedical Research Centre at Guy’s and St. Thomas’ National Health Service, Foundation Trust and King’s College London, UK
| | | | - John Trowsdale
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Sophia Urbanczyk
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Dept. of Internal Medicine III, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Switzerland
| | - Edwin van der Pol
- Vesicle Observation Center; Biomedical Engineering & Physics; Laboratory Experimental Clinical Chemistry; Amsterdam University Medical Centers, Location AMC, The Netherlands
| | - Sofie Van Gassen
- Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | | | - René A.W. van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc Veldhoen
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | | | - Paulo Vieira
- Unit Lymphopoiesis, Department of Immunology, Institut Pasteur, Paris, France
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT) Charité Universitätsmedizin Berlin and Berlin Institute of Health, Core Unit ImmunoCheck
| | - Anouk von Borstel
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | | | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | | | - Paul K. Wallace
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, USA
| | - Sa A. Wang
- Dept of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin M. Wang
- The Scientific Platforms, the Westmead Institute for Medical Research, the Westmead Research Hub, Westmead, New South Wales, Australia
| | | | | | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gary Warnes
- Flow Cytometry Core Facility, Blizard Institute, Queen Mary London University, London, UK
| | - Sarah Warth
- BCRT Flow Cytometry Lab, Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin
| | - Claudia Waskow
- Regeneration in Hematopoiesis, Leibniz-Institute on Aging, Fritz-Lipmann-Institute (FLI), Jena, Germany
- Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | | | - Carsten Watzl
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| | - Leonie Wegener
- Biophysics, R&D Engineering, Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Thomas Weisenburger
- Department of Biology, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Annika Wiedemann
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Dept. Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Germany
| | - Jürgen Wienands
- Institute for Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Anneke Wilharm
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Robert John Wilkinson
- Department of Infectious Disease, Imperial College London, UK
- Wellcome Centre for Infectious Diseases Research in Africa and Department of Medicine, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa
- Tuberculosis Laboratory, The Francis Crick Institute, London, UK
| | - Gerald Willimsky
- Cooperation Unit for Experimental and Translational Cancer Immunology, Institute of Immunology (Charité - Universitätsmedizin Berlin) and German Cancer Research Center (DKFZ), Berlin, Germany
| | - James B. Wing
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Rieke Winkelmann
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Thomas H. Winkler
- Department of Biology, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Oliver F. Wirz
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Alicia Wong
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore
| | - Peter Wurst
- University Bonn, Medical Faculty, Bonn, Germany
| | - Jennie H. M. Yang
- Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institutes of Health Research Biomedical Research Centre at Guy’s and St. Thomas’ National Health Service, Foundation Trust and King’s College London, UK
| | - Juhao Yang
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Alice Yue
- School of Computing Science, Simon Fraser University, Burnaby, Canada
| | - Hanlin Zhang
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Susanne Maria Ziegler
- Department of Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Christina Zielinski
- German Center for Infection Research (DZIF), Munich, Germany
- Institute of Virology, Technical University of Munich, Munich, Germany
- TranslaTUM, Technical University of Munich, Munich, Germany
| | - Jakob Zimmermann
- Maurice Müller Laboratories (Department of Biomedical Research), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, University of Bern, Bern, Switzerland
| | | |
Collapse
|
20
|
Zizzo G, De Santis M, Bosello S, Tolusso B, Alivernini S, De Luca G, Gremese E, Ferraccioli G. Measuring the T-cell down-regulation of TCR-zeta, ZAP-70 and CD28 in arthritis patients: An old tool for new biomarkers. Eur J Immunol 2019; 49:2195-2203. [PMID: 31400229 DOI: 10.1002/eji.201847849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 06/26/2019] [Accepted: 08/08/2019] [Indexed: 11/10/2022]
Abstract
Low T-cell receptor (TCR)/CD28 signaling lymphocytes are expanded in arthritis. We asked whether the down-expression of TCR-related molecules correlates with specific arthritis characteristics and if it has clinical implications. TCR-ZETA, ZAP-70 and CD28 expression was measured by flow cytometry in synovial fluid (SF) and peripheral blood (PB)-derived T cells. In PB, ZETA-downregulation in CD4+ CD28+ and consequent CD4+ CD28lowZETAlow cell expansion correlate with CRP elevation, leukocyte recruitment into SF and, primarily, disease activity (DAS). In some patients, ZETA-downregulation extends to CD8+ CD28null and/or CD8+ CD28+ cells, and this correlates with enhanced leukocyte recruitment, multiple joint involvement, and disability index (HAQ). ZETA-downregulation in CD4+ CD28+ may also lead to CD4+ CD28+ ZETAnull cell expansion, which strongly correlates with HAQ. In SF, ZETA-downregulation in CD8+ CD28null and consequent CD8+ CD28nullZETAlow/null cell expansion correlate with CRP elevation and neutrophilic influx into SF, whereas ZAP-downregulation in CD8+ CD28+ and consequent CD8+ CD28lowZAPlow cell expansion strongly correlate with HAQ and DAS. ZETA-downregulation is preponderant in SF of seronegative arthritides, with seronegative rheumatoid arthritis showing significant down-regulation in CD8+ CD28null, and non-rheumatoid arthritides showing significant down-regulation in CD4+ CD28+ . Altogether, we identified new molecular and cellular biomarkers of arthritis-related T-cell inflammation, useful for assessing arthritis activity, predicting polyarticular progression and functional impairment, characterizing seronegative arthritides, and possibly tailoring immunotherapies.
Collapse
Affiliation(s)
- Gaetano Zizzo
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy.,Rheumatology Unit, Medical Department, Vimercate Hospital, Monza e Brianza, Italy
| | - Maria De Santis
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Silvia Bosello
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - Barbara Tolusso
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - Stefano Alivernini
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Gremese
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - Gianfranco Ferraccioli
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
21
|
Carpentier S, Romagné F, Vivier E. A comprehensive approach to gene expression profiling in immune cells. Methods Enzymol 2019; 636:1-47. [PMID: 32178815 DOI: 10.1016/bs.mie.2019.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
With the advent of whole-transcriptome studies and the growing need for public repositories, it has become essential to combine multiple heterogeneous datasets for immune cells. In this chapter, we describe the implementation of a compendium of 10,833 genes for 975 samples, corresponding to 52 resting immune cell types. We begin by describing the datasets, and their selection, in particular. We then explain the methodology implemented to create a qualified compendium: the processing of each array (quality control, normalization and bias correction), integration (merging rules, global normalization and batch removal) and validation. Finally some examples of use will be detailed. The utility and limitations of the compendium are also discussed, as an introduction to the next version.
Collapse
Affiliation(s)
| | | | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France; Aix-Marseille Univ, APHM, CNRS, INSERM, CIML, Hôpital de la Timone, Marseille-Immunopole, Marseille, France.
| |
Collapse
|
22
|
Molinier-Frenkel V, Prévost-Blondel A, Castellano F. The IL4I1 Enzyme: A New Player in the Immunosuppressive Tumor Microenvironment. Cells 2019; 8:E757. [PMID: 31330829 PMCID: PMC6678094 DOI: 10.3390/cells8070757] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 12/14/2022] Open
Abstract
The high metabolic needs of T lymphocytes in response to activation make them particularly vulnerable to modifications of their biochemical milieu. Immunosuppressive enzymes produced in the tumor microenvironment modify nutrient availability by catabolizing essential or semi-essential amino acids and producing toxic catabolites, thus participating in the local sabotage of the antitumor immune response. L-amino-acid oxidases are FAD-bound enzymes found throughout evolution, from bacteria to mammals, and are often endowed with anti-infectious properties. IL4I1 is a secreted L-phenylalanine oxidase mainly produced by inflammatory antigen-presenting cells-in particular, macrophages present in T helper type 1 granulomas and in various types of tumors. In the last decade, it has been shown that IL4I1 is involved in the fine control of B- and T-cell adaptive immune responses. Preclinical models have revealed its role in cancer immune evasion. Recent clinical data highlight IL4I1 as a new potential prognostic marker in human melanoma. As a secreted enzyme, IL4I1 may represent an easily targetable molecule for cancer immunotherapy.
Collapse
Affiliation(s)
- Valérie Molinier-Frenkel
- INSERM, U955, Team 09, 94010 Créteil, France.
- Faculty of Medicine, University Paris Est, 94010 Créteil, France.
- AP-HP, H. Mondor - A. Chenevier Hospital, Biological Immunology Service, 94010 Créteil, France.
| | - Armelle Prévost-Blondel
- INSERM, U1016, Institute Cochin, 75014 Paris, France
- CNRS, UMR8104, 75014 Paris, France
- University Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Flavia Castellano
- INSERM, U955, Team 09, 94010 Créteil, France.
- Faculty of Medicine, University Paris Est, 94010 Créteil, France.
| |
Collapse
|
23
|
Kunkl M, Mastrogiovanni M, Porciello N, Caristi S, Monteleone E, Arcieri S, Tuosto L. CD28 Individual Signaling Up-regulates Human IL-17A Expression by Promoting the Recruitment of RelA/NF-κB and STAT3 Transcription Factors on the Proximal Promoter. Front Immunol 2019; 10:864. [PMID: 31068940 PMCID: PMC6491678 DOI: 10.3389/fimmu.2019.00864] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/04/2019] [Indexed: 01/22/2023] Open
Abstract
CD28 is an important co-stimulatory receptor for T lymphocytes that, in humans, delivers TCR-independent signal leading to the up-regulation of pro-inflammatory cytokines. We have recently reported that CD28 autonomous signaling induces the expression of IL-17A in peripheral CD4+ T lymphocytes from healthy donors, multiple sclerosis, and type 1 diabetes patients. Due to the relevance of IL-17A in the pathophysiology of several inflammatory and autoimmune diseases, we characterized the mechanisms and signaling mediators responsible for CD28-induced IL-17A expression. Here we show that CD28-mediated up-regulation of IL-17A gene expression depends on RelA/NF-κB and IL-6-associated STAT3 transcriptions factors. In particular, we found that CD28-activated RelA/NF-κB induces the expression of IL-6 that, in a positive feedback loop, mediates the activation and nuclear translocation of tyrosine phosphorylated STAT3 (pSTAT3). pSTAT3 in turn cooperates with RelA/NF-κB by binding specific sequences within the proximal promoter of human IL-17A gene, thus inducing its expression. Finally, by using specific inhibitory drugs, we also identified class 1A phosphatidylinositol 3-kinase (PI3K) as a critical upstream regulator of CD28-mediated RelA/NF-κB and STAT3 recruitments and trans-activation of IL-17A promoter. Our findings reveal a novel mechanism by which human CD28 may amplify IL-17A expression in human T lymphocytes and provide biological bases for immunotherapeutic approaches targeting CD28-associated class 1A PI3K to dampen IL-17A-mediated inflammatory response in autoimmune/inflammatory disorders.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Marta Mastrogiovanni
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Pasteur Institute, Paris, France
| | - Nicla Porciello
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Silvana Caristi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Emanuele Monteleone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Stefano Arcieri
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| |
Collapse
|
24
|
Maggi L, Mazzoni A, Cimaz R, Liotta F, Annunziato F, Cosmi L. Th17 and Th1 Lymphocytes in Oligoarticular Juvenile Idiopathic Arthritis. Front Immunol 2019; 10:450. [PMID: 30930898 PMCID: PMC6428030 DOI: 10.3389/fimmu.2019.00450] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/19/2019] [Indexed: 01/16/2023] Open
Abstract
In the last years much attention has focused on the Th17 and Th1 phenotypes and on their pathogenic role in juvenile idiopathic arthritis, investigating how the cytokines produced by T helper cells act on resident cells on the synovia and which signal transduction pathways regulate Th17 cells proliferation and plasticity. In this context, an important milestone was represented by the identification of the non-classic Th1 phenotype, developed from the shift of Th17 cells. The cytokine TNF-α, beyond its well-known proinflammatory activity is involved in this process and this is one of the reasons why the TNF-α inhibitors are widely used in the treatment of juvenile idiopathic arthritis patients.
Collapse
Affiliation(s)
- Laura Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Florence, Italy
| | - Rolando Cimaz
- Anna Meyer Children's Hospital and University of Florence, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Florence, Italy
| |
Collapse
|
25
|
Ramspott JP, Bekkat F, Bod L, Favier M, Terris B, Salomon A, Djerroudi L, Zaenker KS, Richard Y, Molinier-Frenkel V, Castellano F, Avril MF, Prévost-Blondel A. Emerging Role of IL-4–Induced Gene 1 as a Prognostic Biomarker Affecting the Local T-Cell Response in Human Cutaneous Melanoma. J Invest Dermatol 2018; 138:2625-2634. [DOI: 10.1016/j.jid.2018.06.178] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/04/2018] [Accepted: 06/18/2018] [Indexed: 12/28/2022]
|
26
|
Mazzoni A, Maggi L, Siracusa F, Ramazzotti M, Rossi MC, Santarlasci V, Montaini G, Capone M, Rossettini B, Palma R, Kruglov A, Chang H, Cimaz R, Maggi E, Romagnani S, Liotta F, Cosmi L, Annunziato F. Eomes
controls the development of Th17‐derived (non‐classic) Th1 cells during chronic inflammation. Eur J Immunol 2018; 49:79-95. [DOI: 10.1002/eji.201847677] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/20/2018] [Accepted: 08/20/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | | | - Matteo Ramazzotti
- Department of Biomedical Experimental and Clinical Sciences “Mario Serio” University of Florence Firenze Italy
| | - Maria Caterina Rossi
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Veronica Santarlasci
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Gianni Montaini
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Beatrice Rossettini
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Raffaele Palma
- Diparimento di Medicina di Precisione Università della Campania Napoli Italy
- Institute of Protein Biochemistry CNR Napoli
| | | | | | - Rolando Cimaz
- Anna Meyer Children's Hospital and University of Florence Italy
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine and DENOTHE Center University of Florence Firenze Italy
- Flow cytometry and Immunotherapy Diagnostic Center Azienda Ospedaliera Careggi Florence Italy
| |
Collapse
|
27
|
Shigeno Y, Zhang H, Banno T, Usuda K, Nochi T, Inoue R, Watanabe G, Jin W, Benno Y, Nagaoka K. Gut microbiota development in mice is affected by hydrogen peroxide produced from amino acid metabolism during lactation. FASEB J 2018; 33:3343-3352. [PMID: 30433825 DOI: 10.1096/fj.201801462r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The development of gut microbiota during infancy is an important event that affects the health status of the host; however, the mechanism governing it is not fully understood. l-Amino acid oxidase 1 (LAO1) is a flavoprotein that catalyzes the oxidative deamination of particular l-amino acids and converts them into keto acids, ammonia, and H2O2. Our previous study showed that LAO1 is present in mouse milk and exerts protection against bacteria by its production of H2O2. The data led us to consider whether LAO1, H2O2, or both could impact infant gut microbiota development via mother's milk consumption in mice. Different gut microbiota profiles were observed in the wild-type (WT) and LAO1-knockout mouse pups. The WT pups' microbiota was relatively simple and composed of only a few dominant bacteria, such as Lactobacillus, whereas the lactating knockout pups had high microbiota diversity. Cross-fostering experiments indicated that WT milk (containing LAO1) has the ability to suppress the diversity of microbiota in pups. We observed that the stomach content of pups fed WT milk had LAO1 proteins and the ability to produce H2O2. Moreover, culture experiments showed that Lactobacillus was abundant in the feces of pups fed WT milk and that Lactobacillus was more resistant to H2O2 than Bifidobacterium and Escherichia. Human breast milk produces very little H2O2, which could be the reason for Lactobacillus not being dominant in the feces of breast-fed human infants. In mouse mother's milk, H2O2 is generated from the process of free amino acid metabolism, and H2O2 may be a key player in regulating the initial acquisition and development of gut microbiota, especially growth of Lactobacillus, during infancy.-Shigeno, Y., Zhang, H., Banno, T., Usuda, K., Nochi, T., Inoue, R., Watanabe, G., Jin, W., Benno, Y., Nagaoka, K. Gut microbiota development in mice is affected by hydrogen peroxide produced from amino acid metabolism during lactation.
Collapse
Affiliation(s)
- Yuko Shigeno
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan.,Benno Laboratory, RIKEN Innovation Center, Wako, Japan
| | - Haolin Zhang
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Taihei Banno
- Benno Laboratory, RIKEN Innovation Center, Wako, Japan
| | - Kento Usuda
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tomonori Nochi
- Laboratory of Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Kyoto, Japan; and
| | - Gen Watanabe
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Wanzhu Jin
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yoshimi Benno
- Benno Laboratory, RIKEN Innovation Center, Wako, Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
28
|
Barra G, Lepore A, Gagliardi M, Somma D, Matarazzo MR, Costabile F, Pasquale G, Mazzoni A, Gallo C, Nuzzo G, Annunziato F, Fontana A, Leonardi A, De Palma R. Sphingosine Kinases promote IL-17 expression in human T lymphocytes. Sci Rep 2018; 8:13233. [PMID: 30185808 PMCID: PMC6125344 DOI: 10.1038/s41598-018-31666-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 08/22/2018] [Indexed: 12/31/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) has a role in many cellular processes. S1P is involved in cell growth and apoptosis, regulation of cell trafficking, production of cytokines and chemokines. The kinases SphK1 and SphK2 (SphKs) phosphorilate Sphingosine (Sph) to S1P and several phosphatases revert S1P to sphingosine, thus assuring a balanced pool that can be depleted by a Sphingosine lyase in hexadecenal compounds and aldehydes. There are evidences that SphK1 and 2 may per se control cellular processes. Here, we report that Sph kinases regulate IL-17 expression in human T cells. SphKs inhibition impairs the production of IL-17, while their overexpression up-regulates expression of the cytokine through acetylation of IL-17 promoter. SphKs were up-regulated also in PBMCs of patients affected by IL-17 related diseases. Thus, S1P/S1P kinases axis is a mechanism likely to promote IL-17 expression in human T cells, representing a possible therapeutic target in human inflammatory diseases.
Collapse
Affiliation(s)
- Giusi Barra
- Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Italy
| | - Alessio Lepore
- Univeristy of Naples "Federico II", Department of Molecular Medicine and Medical Biotechnology, Napoli, Italy
| | - Miriam Gagliardi
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso', CNR, Napoli, 80131, Italy
| | - Domenico Somma
- Univeristy of Naples "Federico II", Department of Molecular Medicine and Medical Biotechnology, Napoli, Italy
| | | | - Francesca Costabile
- Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Italy
| | - Giuseppe Pasquale
- Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Carmela Gallo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, Pozzuoli, 80078, Italy
| | - Genoveffa Nuzzo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, Pozzuoli, 80078, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Angelo Fontana
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, Pozzuoli, 80078, Italy
| | - Antonio Leonardi
- Univeristy of Naples "Federico II", Department of Molecular Medicine and Medical Biotechnology, Napoli, Italy
| | - Raffaele De Palma
- Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Italy. .,Institute of Protein Biochemistry-CNR, via P. Castellino, 111, 80131, Napoli, Italy.
| |
Collapse
|
29
|
Chen Z, Chen S, Liu J. The role of T cells in the pathogenesis of Parkinson's disease. Prog Neurobiol 2018; 169:1-23. [PMID: 30114440 DOI: 10.1016/j.pneurobio.2018.08.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/24/2018] [Accepted: 08/12/2018] [Indexed: 02/06/2023]
Abstract
Recent evidence has shown that neuroinflammation plays a key role in the pathogenesis of Parkinson's disease (PD). However, different components of the brain's immune system may exert diverse effects on neuroinflammatory events in PD. The adaptive immune response, especially the T cell response, can trigger type 1 pro-inflammatory activities and suppress type 2 anti-inflammatory activities, eventually resulting in deregulated neuroinflammation and subsequent dopaminergic neurodegeneration. Additionally, studies have increasingly shown that therapies targeting T cells can alleviate neurodegeneration and motor behavior impairment in animal models of PD. Therefore, we conclude that abnormal T cell-mediated immunity is a fundamental pathological process that may be a promising translational therapeutic target for Parkinson's disease.
Collapse
Affiliation(s)
- Zhichun Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated with the Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
30
|
Ohl K, Nickel H, Moncrieffe H, Klemm P, Scheufen A, Föll D, Wixler V, Schippers A, Wagner N, Wedderburn LR, Tenbrock K. The transcription factor CREM drives an inflammatory phenotype of T cells in oligoarticular juvenile idiopathic arthritis. Pediatr Rheumatol Online J 2018; 16:39. [PMID: 29925386 PMCID: PMC6011589 DOI: 10.1186/s12969-018-0253-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Inflammatory effector T cells trigger inflammation despite increased numbers of Treg cells in the synovial joint of patients suffering from juvenile idiopathic arthritis (JIA). The cAMP response element (CREM)α is known to play a major role in regulation of T cells in SLE, colitis, and EAE. However, its role in regulation of effector T cells within the inflammatory joint is unknown. METHODS CREM expression was analyzed in synovial fluid cells from oligoarticular JIA patients by flow cytometry. Peripheral blood mononuclear cells were incubated with synovial fluid and analyzed in the presence and absence of CREM using siRNA experiments for T cell phenotypes. To validate the role of CREM in vivo, ovalbumin-induced T cell dependent arthritis experiments were performed. RESULTS CREM is highly expressed in synovial fluid T cells and its expression can be induced by treating healthy control PBMCs with synovial fluid. Specifically, CREM is more abundant in CD161+ subsets, than CD161- subsets, of T cells and contributes to cytokine expression by these cells. Finally, development of ovalbumin-induced experimental arthritis is ameliorated in mice with adoptively transferred CREM-/- T cells. CONCLUSION In conclusion, our study reveals that beyond its role in SLE T cells CREM also drives an inflammatory phenotype of T cells in JIA.
Collapse
Affiliation(s)
- Kim Ohl
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Helge Nickel
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Halima Moncrieffe
- 0000 0000 9025 8099grid.239573.9Center for Autoimmune Genomics & Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,0000 0001 2179 9593grid.24827.3bDepartment of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH USA
| | - Patricia Klemm
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Anja Scheufen
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Dirk Föll
- 0000 0004 0551 4246grid.16149.3bDepartment of Pediatric Rheumatology and Immunology, University Hospital Muenster, Muenster, Germany
| | - Viktor Wixler
- 0000 0001 2172 9288grid.5949.1Institute of Virology, Westfaelische Wilhelms University, 48149 Muenster, Germany
| | - Angela Schippers
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Norbert Wagner
- 0000 0001 0728 696Xgrid.1957.aDepartment of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Lucy R. Wedderburn
- Arthritis Research UK Centre for Adolescent Rheumatology at UCL UCLH and GOSH, London, UK ,0000000121901201grid.83440.3bUCL GOS Institute of Child Health, University College London, London, UK ,0000 0001 2116 3923grid.451056.3NIHR- Great Ormond Street Hospital Biomedical Research Centre (BRC), London, UK
| | - Klaus Tenbrock
- Department of Pediatrics, Medical Faculty, RWTH Aachen, Pauwelsstr. 30, D-52074, Aachen, Germany.
| |
Collapse
|
31
|
Kang EG, Wu S, Gupta A, von Mackensen YL, Siemetzki H, Freudenberg JM, Wigger-Alberti W, Yamaguchi Y. A phase I randomized controlled trial to evaluate safety and clinical effect of topically applied GSK2981278 ointment in a psoriasis plaque test. Br J Dermatol 2018; 178:1427-1429. [PMID: 29150844 DOI: 10.1111/bjd.16131] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- E G Kang
- GlaxoSmithKline, Collegeville, 1250 S. Collegeville Rd, Collegeville, PA, 19426, U.S.A
| | - S Wu
- GlaxoSmithKline, Collegeville, 1250 S. Collegeville Rd, Collegeville, PA, 19426, U.S.A
| | - A Gupta
- GlaxoSmithKline, Collegeville, 1250 S. Collegeville Rd, Collegeville, PA, 19426, U.S.A
| | | | - H Siemetzki
- bioskin GmbH, Burchardstrasse 17, Hamburg, 20095, Germany
| | - J M Freudenberg
- GlaxoSmithKline, Collegeville, 1250 S. Collegeville Rd, Collegeville, PA, 19426, U.S.A
| | | | - Y Yamaguchi
- GlaxoSmithKline, Collegeville, 1250 S. Collegeville Rd, Collegeville, PA, 19426, U.S.A
| |
Collapse
|
32
|
Bulitta B, Zuschratter W, Bernal I, Bruder D, Klawonn F, von Bergen M, Garritsen HSP, Jänsch L. Proteomic definition of human mucosal-associated invariant T cells determines their unique molecular effector phenotype. Eur J Immunol 2018; 48:1336-1349. [PMID: 29749611 DOI: 10.1002/eji.201747398] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 03/10/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022]
Abstract
Mucosal-associated invariant T cells (MAIT) constitute the most abundant anti-bacterial CD8+ T-cell population in humans. MR1/TCR-activated MAIT cells were reported to organize cytotoxic and innate-like responses but knowledge about their molecular effector phenotype is still fragmentary. Here, we have examined the functional inventory of human MAIT cells (CD3+ Vα7.2+ CD161+ ) in comparison with those from conventional non-MAIT CD8+ T cells (cCD8+ ) and NK cells. Quantitative mass spectrometry characterized 5500 proteins of primary MAIT cells and identified 160 and 135 proteins that discriminate them from cCD8+ T cells and NK cells donor-independently. Most notably, MAIT cells showed a unique exocytosis machinery in parallel to a proinflammatory granzyme profile with high levels of the granzymes A, K, and M. Furthermore, 24 proteins were identified with highest abundances in MAIT cells, including CD26, CD98, and L-amino-oxidase (LAAO). Among those, expression of granzyme K and CD98 were validated as MAIT-specific with respect to non-MAIT CD8+ effector subsets and LAAO was found to be recruited together with granzymes, perforin, and CD107a at the immunological synapse of activated MAIT cells. In conclusion, this study complements knowledge on the molecular effector phenotype of MAIT cells and suggest novel immune regulatory functions as part of their cytotoxic responses.
Collapse
Affiliation(s)
- Björn Bulitta
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Werner Zuschratter
- Special Lab Electron and Laserscanning Microscopy, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Isabel Bernal
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Medical Microbiology and Hospital Hygiene, University Hospital Magdeburg, Magdeburg, Germany
| | - Dunja Bruder
- Institute of Medical Microbiology and Hospital Hygiene, University Hospital Magdeburg, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Frank Klawonn
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Computer Science, Ostfalia University of Applied Sciences, Wolfenbüttel, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Henrikus Stephanus Paulus Garritsen
- Institute for Clinical Transfusion Medicine, Städtisches Klinikum Braunschweig, Braunschweig, Germany.,Fraunhofer Institute for Surface Engineering and Thin Films, Braunschweig, Germany
| | - Lothar Jänsch
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
33
|
van Loosdregt J, van Wijk F, Prakken B, Vastert B. Update on research and clinical translation on specific clinical areas from biology to bedside: Unpacking the mysteries of juvenile idiopathic arthritis pathogenesis. Best Pract Res Clin Rheumatol 2018; 31:460-475. [PMID: 29773267 DOI: 10.1016/j.berh.2018.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 02/08/2023]
Abstract
In the past decades, we have gained important insights into the mechanisms of disease and therapy underlying chronic inflammation in juvenile idiopathic arthritis (JIA). These insights have resulted in several game-changing therapeutic modalities for many patients. However, additional progress still has to be made with regard to efficacy, cost reduction, minimization of side effects, and dose-tapering and stop strategies of maintenance drugs. Moreover, to really transform the current therapeutic strategies into personalized medicine, we need validated biomarkers to translate increased insights into clinical practice. In this article, we describe recent developments in JIA research and outline how clinical innovations need to go hand in hand with basic discoveries to really effect care for patients. Facilitating the transition from bench to bedside is crucial for addressing the major current challenges in JIA management. When successful, it will set new standards for a safe, targeted, and personalized medicine in JIA.
Collapse
Affiliation(s)
- Jorg van Loosdregt
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Femke van Wijk
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Berent Prakken
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Bas Vastert
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands.
| |
Collapse
|
34
|
Bod L, Douguet L, Auffray C, Lengagne R, Bekkat F, Rondeau E, Molinier-Frenkel V, Castellano F, Richard Y, Prévost-Blondel A. IL-4-Induced Gene 1: A Negative Immune Checkpoint Controlling B Cell Differentiation and Activation. THE JOURNAL OF IMMUNOLOGY 2017; 200:1027-1038. [PMID: 29288206 DOI: 10.4049/jimmunol.1601609] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/26/2017] [Indexed: 11/19/2022]
Abstract
Emerging data highlight the crucial role of enzymes involved in amino acid metabolism in immune cell biology. IL-4-induced gene-1 (IL4I1), a secreted l-phenylalanine oxidase expressed by APCs, has been detected in B cells, yet its immunoregulatory role has only been explored on T cells. In this study, we show that IL4I1 regulates multiple steps in B cell physiology. Indeed, IL4I1 knockout mice exhibit an accelerated B cell egress from the bone marrow, resulting in the accumulation of peripheral follicular B cells. They also present a higher serum level of natural Igs and self-reactive Abs. We also demonstrate that IL4I1 produced by B cells themselves controls the germinal center reaction, plasma cell differentiation, and specific Ab production in response to T dependent Ags, SRBC, and NP-KLH. In vitro, IL4I1-deficient B cells proliferate more efficiently than their wild-type counterparts in response to BCR cross-linking. Moreover, the absence of IL4I1 increases activation of the Syk-Akt-S6kinase signaling pathway and calcium mobilization, and inhibits SHP-1 activity upon BCR engagement, thus supporting that IL4I1 negatively controls BCR-dependent activation. Overall, our study reveals a new perspective on IL4I1 as a key regulator of B cell biology.
Collapse
Affiliation(s)
- Lloyd Bod
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Laetitia Douguet
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Cédric Auffray
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Renée Lengagne
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Fériel Bekkat
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Elena Rondeau
- INSERM, U1016, Institut Cochin, 75014 Paris, France.,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09, 94000 Créteil, France.,Faculté de Médecine, Université Paris Est, 94000 Créteil, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Henri Mondor - Albert Chenevier, Service d'Immunologie Biologique, 94000 Créteil, France; and
| | - Flavia Castellano
- INSERM, U955, Equipe 09, 94000 Créteil, France.,Faculté de Médecine, Université Paris Est, 94000 Créteil, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Henri Mondor - Albert Chenevier, Plateforme de Ressources Biologiques, 94000 Créteil, France
| | - Yolande Richard
- INSERM, U1016, Institut Cochin, 75014 Paris, France; .,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Armelle Prévost-Blondel
- INSERM, U1016, Institut Cochin, 75014 Paris, France; .,CNRS, UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| |
Collapse
|
35
|
An Overview of l-Amino Acid Oxidase Functions from Bacteria to Mammals: Focus on the Immunoregulatory Phenylalanine Oxidase IL4I1. Molecules 2017; 22:molecules22122151. [PMID: 29206151 PMCID: PMC6149928 DOI: 10.3390/molecules22122151] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/23/2017] [Accepted: 11/29/2017] [Indexed: 01/04/2023] Open
Abstract
l-amino acid oxidases are flavin adenine dinucleotide-dependent enzymes present in all major kingdom of life, from bacteria to mammals. They participate in defense mechanisms by limiting the growth of most bacteria and parasites. A few mammalian LAAOs have been described, of which the enzyme “interleukin-4 induced gene 1” (IL4I1) is the best characterized. IL4I1 mainly oxidizes l-phenylalanine. It is a secreted enzyme physiologically produced by antigen presenting cells of the myeloid and B cell lineages and T helper type (Th) 17 cells. Important roles of IL4I1 in the fine control of the adaptive immune response in mice and humans have emerged during the last few years. Indeed, IL4I1 inhibits T cell proliferation and cytokine production and facilitates naïve CD4+ T-cell differentiation into regulatory T cells in vitro by limiting the capacity of T lymphocytes to respond to clonal receptor stimulation. It may also play a role in controlling the germinal center reaction for antibody production and limiting Th1 and Th17 responses. IL4I1 is expressed in tumor-associated macrophages of most human cancers and in some tumor cell types. Such expression, associated with its capacity to facilitate tumor growth by inhibiting the anti-tumor T-cell response, makes IL4I1 a new potential druggable target in the field of immunomodulation in cancer.
Collapse
|
36
|
The Th17 Lineage: From Barrier Surfaces Homeostasis to Autoimmunity, Cancer, and HIV-1 Pathogenesis. Viruses 2017; 9:v9100303. [PMID: 29048384 PMCID: PMC5691654 DOI: 10.3390/v9100303] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022] Open
Abstract
The T helper 17 (Th17) cells represent a subset of CD4+ T-cells with unique effector functions, developmental plasticity, and stem-cell features. Th17 cells bridge innate and adaptive immunity against fungal and bacterial infections at skin and mucosal barrier surfaces. Although Th17 cells have been extensively studied in the context of autoimmunity, their role in various other pathologies is underexplored and remains an area of open investigation. This review summarizes the history of Th17 cell discovery and the current knowledge relative to the beneficial role of Th17 cells in maintaining mucosal immunity homeostasis. We further discuss the concept of Th17 pathogenicity in the context of autoimmunity, cancer, and HIV infection, and we review the most recent discoveries on molecular mechanisms regulating HIV replication/persistence in pathogenic Th17 cells. Finally, we stress the need for novel fundamental research discovery-based Th17-specific therapeutic interventions to treat pathogenic conditions associated with Th17 abnormalities, including HIV infection.
Collapse
|
37
|
Aubatin A, Sako N, Decrouy X, Donnadieu E, Molinier-Frenkel V, Castellano F. IL4-induced gene 1 is secreted at the immune synapse and modulates TCR activation independently of its enzymatic activity. Eur J Immunol 2017; 48:106-119. [PMID: 28891065 DOI: 10.1002/eji.201646769] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 08/21/2017] [Accepted: 09/06/2017] [Indexed: 01/11/2023]
Abstract
Amino-acid catabolizing enzymes produced by mononuclear phagocytes play a central role in regulating the immune response. The mammalian phenylalanine-catabolizing enzyme IL4-induced gene 1 (IL4I1) inhibits effector T lymphocyte proliferation and facilitates regulatory T-cell development. IL4I1 expression by macrophages of various human tumors may affect patient prognosis as it facilitates tumor escape from the T-cell response in murine models. Its enzymatic activity appears to participate in its effects, but some actions of IL4I1 remain unclear. Here, we show that the presence of IL4I1 during T-cell activation decreases early signaling events downstream of TCR stimulation, resulting in global T-cell inhibition which is more pronounced when there is CD28 costimulation. Surprisingly, the enzymatic activity of IL4I1 is not involved. Focal secretion of IL4I1 into the immune synaptic cleft and its binding to CD3+ lymphocytes could be important in IL4I1 immunosuppressive mechanism of action.
Collapse
Affiliation(s)
- Aude Aubatin
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Nouhoum Sako
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Xavier Decrouy
- INSERM, U955, Plateforme d'imagerie, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France
| | - Emmanuel Donnadieu
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Service d'Immunologie Biologique, Créteil, France
| | - Flavia Castellano
- INSERM, U955, Equipe 09, Créteil, France.,Faculté de Médecine, Université Paris Est, Créteil, France.,AP-HP, Hôpital H. Mondor - A. Chenevier, Plateforme de Ressources Biologiques, Créteil, France
| |
Collapse
|
38
|
Molinier-Frenkel V, Castellano F. Immunosuppressive enzymes in the tumor microenvironment. FEBS Lett 2017; 591:3135-3157. [DOI: 10.1002/1873-3468.12784] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/31/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Valérie Molinier-Frenkel
- INSERM, U955, Equipe 09; Créteil France
- Faculté de Médecine; Université Paris Est; Créteil France
- Service d'Immunologie Biologique; AP-HP, Hôpital H. Mondor - A. Chenevier; Créteil France
| | - Flavia Castellano
- INSERM, U955, Equipe 09; Créteil France
- Faculté de Médecine; Université Paris Est; Créteil France
- Plateforme de Ressources Biologiques; AP-HP, Hôpital H. Mondor - A. Chenevier; Créteil France
| |
Collapse
|
39
|
Santarlasci V, Mazzoni A, Capone M, Rossi MC, Maggi L, Montaini G, Rossettini B, Cimaz R, Ramazzotti M, Barra G, De Palma R, Maggi E, Liotta F, Cosmi L, Romagnani S, Annunziato F. Musculin inhibits human T-helper 17 cell response to interleukin 2 by controlling STAT5B activity. Eur J Immunol 2017; 47:1427-1442. [PMID: 28612433 DOI: 10.1002/eji.201746996] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/22/2017] [Accepted: 06/09/2017] [Indexed: 01/01/2023]
Abstract
We recently demonstrated that human T-helper (Th) 17 cells, unlike Th1 cells, do not proliferate in response to T-cell receptor stimulation, mainly because of their reduced capacity to produce and respond to IL-2. In this study, we show that their lower responsiveness to IL-2 is due to the selective expression of Musculin (MSC), a member of the basic helix-loop-helix transcription factors. We show that MSC expression in human Th17 cells is retinoic acid orphan receptor (ROR)γt-dependent, and allows the upregulation of PPP2R2B, a regulatory member of the protein phosphatase 2A (PP2A) enzyme. High PPP2R2B levels in human Th17 cells were responsible for the reduced STAT5B Ser-193 phosphorylation upon IL-2 signalling and, therefore, impaired STAT5B DNA binding and transcriptional activity on IL-2 target genes. PP2A, observed in Th17 cells, controls also STAT3, dephosphorylating Ser727, thus increasing its activity that plays a crucial role in Th17 development and/or maintenance. Thus, our findings identify an additional mechanism responsible for the limited expansion of human Th17 cells, and could provide a further explanation for the rarity of these cells in inflamed tissues.
Collapse
Affiliation(s)
- Veronica Santarlasci
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Maria Caterina Rossi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Gianni Montaini
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Beatrice Rossettini
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Rolando Cimaz
- Anna Meyer Children's Hospital and University of Florence, Italy
| | - Matteo Ramazzotti
- Department of Biomedical Experimental and Clinical Sciences "Mario Serio" University of Florence, Firenze, Italy
| | - Giusi Barra
- Department of Clinical and Experimental Medicine, Università della Campania "L. Vanvitelli,", Napoli, Italy
| | - Raffaele De Palma
- Department of Clinical and Experimental Medicine, Università della Campania "L. Vanvitelli,", Napoli, Italy.,Institute of Protein Biochemistry, CNR, Napoli
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, Firenze, Italy.,Regenerative Medicine Unit and Immunology and Cellular Therapy Unit of Azienda Ospedaliera Careggi, Florence, Italy
| |
Collapse
|
40
|
Romagnani S. IL4I1: Key immunoregulator at a crossroads of divergent T-cell functions. Eur J Immunol 2017; 46:2302-2305. [PMID: 27726138 DOI: 10.1002/eji.201646617] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 08/04/2016] [Accepted: 08/24/2016] [Indexed: 01/17/2023]
Abstract
The interleukin (IL)-4-induced gene1 (IL4I1), which encodes the L-amino acid oxidase enzyme, plays an important immunoregulatory role. Indeed, this enzyme which is produced by B cells-including neoplastic B cells-dendritic cells and macrophages has been shown to inhibit proliferation, cytotoxicity and IFN-γ production by tumor-infiltrating CD8+ T cells, thus favoring tumor escape. Moreover, the same gene has been found to be constitutively expressed by CD4+ T helper 17 (Th17) cells, where it down-regulates cell proliferation through a reduction of CD3 chains expression in the T-cell receptor complex, thus impairing IL-2 production, and by maintaining in the same cells a high expression of Tob1, which inhibits cell cycle entry, through a still unknown mechanism. Finally, IL4I1 has been shown to drive the differentiation of naive T cells into inducible regulatory T (iTreg) cells. Taken together, IL4I1 down-regulates the effector CD8+ T-cell response, promotes the development of iTreg cells and limits the expansion of Th17 cells, thus not only favoring tumor escape, but also reducing the potentially dangerous effects of adaptive immune responses in chronic inflammatory disorders.
Collapse
Affiliation(s)
- Sergio Romagnani
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy.
| |
Collapse
|
41
|
Kunkl M, Porciello N, Mastrogiovanni M, Capuano C, Lucantoni F, Moretti C, Persson JL, Galandrini R, Buzzetti R, Tuosto L. ISA-2011B, a Phosphatidylinositol 4-Phosphate 5-Kinase α Inhibitor, Impairs CD28-Dependent Costimulatory and Pro-inflammatory Signals in Human T Lymphocytes. Front Immunol 2017; 8:502. [PMID: 28491063 PMCID: PMC5405084 DOI: 10.3389/fimmu.2017.00502] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/12/2017] [Indexed: 12/16/2022] Open
Abstract
Phosphatidylinositol 4,5-biphosphate (PIP2) is a membrane phospholipid that controls the activity of several proteins regulating cytoskeleton reorganization, cytokine gene expression, T cell survival, proliferation, and differentiation. Phosphatidylinositol 4-phosphate 5-kinases (PIP5Ks) are the main enzymes involved in PIP2 biosynthesis by phosphorylating phosphatidylinositol 4-monophosphate (PI4P) at the D5 position of the inositol ring. In human T lymphocytes, we recently found that CD28 costimulatory molecule is pivotal for PIP2 turnover by recruiting and activating PIP5Kα. We also found that PIP5Kα is the main regulator of both CD28 costimulatory signals integrating those delivered by TCR as well as CD28 autonomous signals regulating the expression of pro-inflammatory genes. Given emerging studies linking alterations of PIP2 metabolism to immune-based diseases, PIP5Kα may represent a promising target to modulate immunity and inflammation. Herewith, we characterized a recently discovered inhibitor of PIP5Kα, ISA-2011B, for its inhibitory effects on T lymphocyte functions. We found that the inhibition of PIP5Kα lipid-kinase activity by ISA-2011B significantly impaired CD28 costimulatory signals necessary for TCR-mediated Ca2+ influx, NF-AT transcriptional activity, and IL-2 gene expression as well as CD28 autonomous signals regulating the activation of NF-κB and the transcription of pro-inflammatory cytokine and chemokine genes. Moreover, our data on the inhibitory effects of ISA-2011B on CD28-mediated upregulation of inflammatory cytokines related to Th17 cell phenotype in type 1 diabetes patients suggest ISA-2011B as a promising anti-inflammatory drug.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Nicla Porciello
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Marta Mastrogiovanni
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Cristina Capuano
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Chiara Moretti
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Jenny L Persson
- Division of Experimental Cancer Research, Department of Laboratory Medicine, Clinical Research Center, Lund University, Malmö, Sweden.,Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | | | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
42
|
Nanke Y, Kobashigawa T, Yago T, Kawamoto M, Yamanaka H, Kotake S. Detection of IFN-γ+IL-17+ cells in salivary glands of patients with Sjögren's syndrome and Mikulicz's disease: Potential role of Th17•Th1 in the pathogenesis of autoimmune diseases. ACTA ACUST UNITED AC 2017; 39:473-477. [PMID: 27795505 DOI: 10.2177/jsci.39.473] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective: Th17 cells, which mainly produce interleukin (IL)-17, have been suggested to play a critical role in the pathogenesis of autoimmune diseases. The plasticity of Th17 cells, in which these cells shift to a Th1 phenotype in the presence of IL-12, has recently been reported. However, the role of IL-17 in Sjögren's syndrome (SS) and Mikulicz's disease (MD) currently remains unknown. PATIENTS AND METHODS The submandibular salivary gland and lymph node of a MD patient and the salivary glands of 15 SS patients were collected. IFN-γ+ cells, IL-17+ cells, and IFN-γ+IL-17+ cells were detected by immunohistochemical staining. RESULTS IFN-γ+ cells, IL-17+ cells, and IFN-γ+IL-17+ cells were detected in the submandibular salivary gland and lymph node of the MD patient and salivary glands of the 15 SS patients. DISCUSSION IFN-γ+IL-17+cells in the salivary glands of patients were speculated to be Th1/Th17 cells in the present study. Th1/Th17 cells are known to be derived from Th17 cells and differentiate into Th1 cells, and IL-17-derived Th1 cells have been suggested to induce the deterioration of juvenile idiopathic arthritis (JIA). Thus, Th1/Th17 cells may play an important role in the pathogenesis of SS and MD. CONCLUSION IFN-γ+, IFN-γ+IL-17+, and IL-17+ cells were detected in the submandibular salivary gland and lymph node of a MD patient and the salivary glands of 15 SS patients.
Collapse
Affiliation(s)
- Yuki Nanke
- Institute of Rheumatology, Tokyo Women's Medical University
| | | | | | | | | | | |
Collapse
|
43
|
Berker M, Frank LJ, Geßner AL, Grassl N, Holtermann AV, Höppner S, Kraef C, Leclaire MD, Maier P, Messerer DAC, Möhrmann L, Nieke JP, Schoch D, Soll D, Woopen CMP. Allergies - A T cells perspective in the era beyond the T H1/T H2 paradigm. Clin Immunol 2016; 174:73-83. [PMID: 27847316 DOI: 10.1016/j.clim.2016.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 11/02/2016] [Accepted: 11/09/2016] [Indexed: 12/11/2022]
Abstract
Allergic diseases have emerged as a major health care burden, especially in the western hemisphere. They are defined by overshooting reactions of an aberrant immune system to harmless exogenous stimuli. The TH1/TH2 paradigm assumes that a dominance of TH2 cell activation and an inadequate TH1 cell response are responsible for the development of allergies. However, the characterization of additional T helper cell subpopulations such as TH9, TH17, TH22, THGM-CSF and their interplay with regulatory T cells suggest further layers of complexity. This review summarizes state-of-the-art knowledge on T cell diversity and their induction, while revisiting the TH1/TH2 paradigm. With respect to these numerous contributors, it offers a new perspective on the pathogenesis of asthma, allergic rhinitis (AR) and atopic dermatitis (AD) incorporating recent discoveries in the field of T cell plasticity.
Collapse
Affiliation(s)
- Moritz Berker
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Larissa Johanna Frank
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Anja Lidwina Geßner
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Niklas Grassl
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Anne Verena Holtermann
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Stefanie Höppner
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Christian Kraef
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany.
| | - Martin Dominik Leclaire
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Pia Maier
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | | | - Lino Möhrmann
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Jan Philipp Nieke
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Diana Schoch
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | - Dominik Soll
- German Academic Scholarship Foundation - Studienstiftung des deutschen Volkes, Bonn, Germany
| | | |
Collapse
|
44
|
Capone M, Maggi L, Santarlasci V, Rossi MC, Mazzoni A, Montaini G, Cimaz R, Ramazzotti M, Piccinni MP, Barra G, De Palma R, Liotta F, Maggi E, Romagnani S, Annunziato F, Cosmi L. Chitinase 3-like-1 is produced by human Th17 cells and correlates with the level of inflammation in juvenile idiopathic arthritis patients. Clin Mol Allergy 2016; 14:16. [PMID: 27826220 PMCID: PMC5100333 DOI: 10.1186/s12948-016-0053-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/25/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND CHI3L1 is a chitinase-like protein without enzymatic activity, produced by activated macrophages, chondrocytes, neutrophils. Recent studies on arthritis, asthma, and inflammatory bowel diseases suggest that chitinases are important in inflammatory processes and tissue remodeling, but their production by human T cells, has never been reported. METHODS A microarray analysis of gene expression profile was performed on Th17 and classic Th1 cell clones and CHI3L1 was found among the up-regulated genes on Th17 cells. Different types of helper T cell clones (TCCs) were then evaluated by Real Time PCR (RT-PCR) for CHI3L1 mRNA expression; protein expression was investigated in cell lysates by western blotting and in cultures supernatants by ELISA. ELISA was also used to measure CHI3L1 in the serum and in the synovial fluid (SF) of juvenile idiopathic arthritis (JIA) patients. RESULTS At mRNA level CHI3L1 was highly expressed by Th17, Th17/Th1, non classic Th1 and even in Th17/Th2 cell clones, whereas it was virtually absent in CD161- classic Th1 and Th2 TCCs. CHI3L1 was also detected in cell culture supernatants of Th17 and Th17-derived cells but not of classic Th1. Moreover CHI3L1 was higher in the SF than in serum of JIA patients, and it positively correlated with the frequency of Th17 and non-classic Th1 cells in SF. CHI3L1 in SF also positively correlated with the C reactive protein (CRP) serum levels, and with the levels of some proinflammatory cytokines, such as IL-6 and p40, which is the common subunit of IL12 and IL23. CONCLUSIONS Here we describe for the first time CHI3L1 production by T cells owing the Th17 family. Moreover the positive correlation found between the frequency of Th17 and Th17-derived cell subsets and CHI3L1 levels in SF of JIA patients, in agreement with the suggested role of these cells in inflammatory process, candidates CHI3L1 as a possible biological target in JIA treatment.
Collapse
Affiliation(s)
- Manuela Capone
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Veronica Santarlasci
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Maria Caterina Rossi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Gianni Montaini
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Rolando Cimaz
- Anna Meyer Children's Hospital and University of Florence, 50134, Florence, Italy
| | - Matteo Ramazzotti
- Dept. of Biomedical Experimental and Clinical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Marie Pierre Piccinni
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Giusi Barra
- Dept. of Clinical & Experimental Medicine, Second University of Naples, 80131 Naples, Italy
| | - Raffaele De Palma
- Dept. of Clinical & Experimental Medicine, Second University of Naples, 80131 Naples, Italy ; Institute of Protein Biochemistry, CNR, 80131, Naples, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Sergio Romagnani
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy ; Department of Internal Medicine, University of Florence, Viale Pieraccini 6, 50134 Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine and DENOTHE Center, University of Florence, 50134 Florence, Italy
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Th17 lymphocytes are now widely believed to be critical for the regulation of various chronic immune diseases, including asthma and chronic obstructive pulmonary disease. In this review, we discuss the current understanding of the role of Th17 cells in the pathogenesis of different asthma phenotypes and chronic obstructive pulmonary disease. RECENT FINDINGS It has been recently reported that Th17 cells and also a new population of Th17/Th2 cells accumulate in bronchoalveolar lavage fluid of asthmatic patients, and positively correlated with airway obstruction and steroid resistance. These patients often have steroid resistant severe asthma and a predominant bronchial neutrophilic inflammation. SUMMARY Steroid resistant severe asthma with predominant bronchial neutrophilic inflammation could benefit from IL-17 targeted therapies. In this view, the definition of clinical phenotypes and inflammatory endotypes of asthma in each patient will be necessary for personalizing the therapeutic approach.
Collapse
|
46
|
Maggi L, Cosmi L, Simonini G, Annunziato F, Cimaz R. T cell subpopulations in juvenile idiopathic arthritis and their modifications after biotherapies. Autoimmun Rev 2016; 15:1141-1144. [PMID: 27640318 DOI: 10.1016/j.autrev.2016.09.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Inflammatory T cells are thought to be central to the pathogenesis of juvenile idiopathic arthritis. In particular, recent evidence has underlined the importance of a balance between Th17 and Treg cells. Several mechanisms have come to light that control this reciprocal relationship. Moreover, it has been shown that in certain conditions, Th17 cells can shift toward a nonclassic Th1 phenotype. Anti-rheumatic biologic therapies may interfere with these mechanisms and re-establish immune tolerance.
Collapse
Affiliation(s)
- Laura Maggi
- Regenerative Medicine Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gabriele Simonini
- Department of Pediatrics, Rheumatology Unit, Anna Meyer Children's Hospital and University of Florence, Italy
| | - Francesco Annunziato
- Regenerative Medicine Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rolando Cimaz
- Department of Pediatrics, Rheumatology Unit, Anna Meyer Children's Hospital and University of Florence, Italy.
| |
Collapse
|
47
|
Wacleche VS, Goulet JP, Gosselin A, Monteiro P, Soudeyns H, Fromentin R, Jenabian MA, Vartanian S, Deeks SG, Chomont N, Routy JP, Ancuta P. New insights into the heterogeneity of Th17 subsets contributing to HIV-1 persistence during antiretroviral therapy. Retrovirology 2016; 13:59. [PMID: 27553844 PMCID: PMC4995622 DOI: 10.1186/s12977-016-0293-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 08/11/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Th17 cells are permissive to HIV-1 infection and their depletion from the gut of infected individuals leads to microbial translocation, a major cause for non-AIDS co-morbidities. Most recent evidence supports the contribution of long-lived Th17 cells to HIV persistence during antiretroviral therapy (ART). However, the identity of long-lived Th17 cells remains unknown. RESULTS Here, we performed an in-depth transcriptional and functional characterization of four distinct Th17 subsets and investigated their contribution to HIV reservoir persistence during ART. In addition to the previously characterized CCR6(+)CCR4(+) (Th17) and CCR6(+)CXCR3(+) (Th1Th17) subsets, we reveal the existence of two novel CCR6(+) subsets, lacking (double negative, CCR6(+)DN) or co-expressing CXCR3 and CCR4 (double positive, CCR6(+)DP). The four subsets shared multiple Th17-polarization markers, a fraction of cells proliferated in response to C. albicans, and exhibited lineage commitment and plasticity when cultured under Th17 and Th1 conditions, respectively. Of note, fractions of CCR6(+)DN and Th17 demonstrated stable Th17-lineage commitment under Th1-polarization conditions. Among the four subsets, CCR6(+)DN expressed a unique transcriptional signature indicative of early Th17 development (IL-17F, STAT3), lymph-node homing (CCR7, CD62L), follicular help (CXCR5, BCL6, ASCL2), and self-renewal (LEFI, MYC, TERC). Cross sectional and longitudinal studies demonstrated that CCR6(+)DN cells were the most predominant CCR6(+) subset in the blood before and after ART initiation; high frequencies of these cells were similarly observed in inguinal lymph nodes of individuals receiving long-term ART. Importantly, replication competent HIV was isolated from CCR6(+)DN of ART-treated individuals. CONCLUSIONS Together, these results provide new insights into the functional heterogeneity of Th17-polarized CCR6(+)CD4(+) T-cells and support the major contribution of CCR6(+)DN cells to HIV persistence during ART.
Collapse
Affiliation(s)
- Vanessa Sue Wacleche
- Département of Microbiologie, Infectiologie Et immunologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada.,Centre de recherche du CHUM, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QC, H2X 0A9, Canada
| | | | - Annie Gosselin
- Centre de recherche du CHUM, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QC, H2X 0A9, Canada
| | - Patricia Monteiro
- Département of Microbiologie, Infectiologie Et immunologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada.,Centre de recherche du CHUM, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QC, H2X 0A9, Canada
| | - Hugo Soudeyns
- Département of Microbiologie, Infectiologie Et immunologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada.,Unité d'immunopathologie virale, Centre de recherche du CHU Sainte-Justine, Montreal, QC, Canada
| | - Rémi Fromentin
- Centre de recherche du CHUM, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QC, H2X 0A9, Canada
| | - Mohammad-Ali Jenabian
- Département des Sciences Biologiques, Université du Québec à Montréal, Montreal, QC, Canada
| | - Shant Vartanian
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Steven G Deeks
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nicolas Chomont
- Département of Microbiologie, Infectiologie Et immunologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada.,Centre de recherche du CHUM, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QC, H2X 0A9, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illness Service and Research Institute, McGill University Health Centre, Montreal, QC, Canada.,Division of Hematology, McGill University Health Centre, Montreal, QC, Canada
| | - Petronela Ancuta
- Département of Microbiologie, Infectiologie Et immunologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada. .,Centre de recherche du CHUM, 900 rue Saint-Denis, Tour Viger, R09.416, Montreal, QC, H2X 0A9, Canada.
| |
Collapse
|
48
|
Serafini B, Rosicarelli B, Veroni C, Zhou L, Reali C, Aloisi F. RORγt Expression and Lymphoid Neogenesis in the Brain of Patients with Secondary Progressive Multiple Sclerosis. J Neuropathol Exp Neurol 2016; 75:877-88. [PMID: 27413074 DOI: 10.1093/jnen/nlw063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 12/16/2022] Open
Abstract
Ectopic B-cell follicle-like structures (ELS) are found in the meninges of patients with secondary progressive multiple sclerosis (SPMS). Because cells expressing the transcriptional regulator retinoic acid receptor-related orphan receptor-γt (RORγt) and producing interleukin 17 (IL17), e.g. T helper 17 cells and lymphoid tissue inducer (LTi) cells, have been implicated in the formation of ELS, we studied RORγt and IL17 expression in brain tissue from patients with SPMS an assessed their relationships to immune infiltrates and meningeal ELS. By immunohistochemistry, small numbers of RORγt-positive cells were detected in the meninges of 6 of 12 SPMS cases analyzed. RORγt-positive cells were localized in B-cell follicles or aggregates and nearby diffuse meningeal infiltrates, and predominantly co-expressed CD3. Only a few RORγt-positive, CD3-negative cells were observed, suggesting the presence of group 3 innate lymphoid cells, which comprise the LTi cell subset. Some IL17-positive cells, co-expressing in part RORγt and predominantly CD3, were found in meningeal B-cell follicles from 4 SPMS cases. Rare RORγt-positive and IL17-positive cells were detected in white matter. Gene expression analysis of laser dissected meningeal infiltrates and white matter lesions confirmed low frequencies and virtual absence of RORγt and IL17 signals, respectively. Thus, there is selective migration or survival of RORγt-positive cells in MS patient meninges and an association of these cells with ELS.
Collapse
Affiliation(s)
- Barbara Serafini
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Barbara Rosicarelli
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Caterina Veroni
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Ling Zhou
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Camilla Reali
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Francesca Aloisi
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR).
| |
Collapse
|
49
|
Murray PJ. Amino acid auxotrophy as a system of immunological control nodes. Nat Immunol 2016; 17:132-9. [PMID: 26784254 PMCID: PMC4893777 DOI: 10.1038/ni.3323] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/13/2015] [Indexed: 02/07/2023]
Abstract
Cells of the immune system are auxotrophs for most amino acids, including several nonessential ones. Arginine and tryptophan are used within the regulatory immune networks to control proliferation and function through pathways that actively deplete the amino acid from the microenvironment or that create regulatory molecules such as nitric oxide or kynurenines. How immune cells integrate information about essential amino acid supplies and then transfer these signals to growth and activation pathways remains unclear but has potential for pathway discovery about amino sensing. In applied research, strategies to harness amino acid auxotrophy so as to block cancerous lymphocyte growth have been attempted for decades with limited success. Emerging insights about amino acid metabolism may lead to new strategies in clinical medicine whereby both amino acid auxotrophy and the immunoregulatory pathways controlled by amino acids can be manipulated.
Collapse
Affiliation(s)
- Peter J Murray
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
50
|
Abstract
Cells of the immune system are auxotrophs for most amino acids, including several nonessential ones. Arginine and tryptophan are used within the regulatory immune networks to control proliferation and function through pathways that actively deplete the amino acid from the microenvironment or that create regulatory molecules such as nitric oxide or kynurenines. How immune cells integrate information about essential amino acid supplies and then transfer these signals to growth and activation pathways remains unclear but has potential for pathway discovery about amino sensing. In applied research, strategies to harness amino acid auxotrophy so as to block cancerous lymphocyte growth have been attempted for decades with limited success. Emerging insights about amino acid metabolism may lead to new strategies in clinical medicine whereby both amino acid auxotrophy and the immunoregulatory pathways controlled by amino acids can be manipulated.
Collapse
|