1
|
Pu D, Wang P, Wang X, Tian Y, Gong H, Ma X, Li M, Zhang D. Focusing on non-responders to infliximab with ulcerative colitis, what can we do first and next? Int Immunopharmacol 2024; 141:112943. [PMID: 39191122 DOI: 10.1016/j.intimp.2024.112943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic immune-mediated inflammation of the colorectum, for which infliximab (IFX) is currently the mainstay of treatment. However, one-third of patients with UC still fail to benefit from the IFX therapy, and early exposure to IFX impairs the efficacy of other subsequent biologics. Therefore, personalized therapeutic system is urgently needed to assist in clinical decision-making and precision treatment. METHODS Four microarray datasets of colonic biopsies from UC patients treated with IFX were obtained from the GEO database to form the Training Cohort and Validation Cohort. Differentially expressed genes (DEGs) in Training Cohort were identified and enriched for GO, KEGG and immune cell infiltration analysis. A prediction model for IFX efficacy was developed based on the LASSO and Logistic regression. The predictive accuracy of the model was verified by the Validation Cohort, and the model-genes/proteins were validated by immunohistochemistry. Gene-drug, gene-ncRNA interaction analysis were performed to identify drugs or non-coding RNAs (ncRNAs) that potentially interacted with the model-genes. Homology Modeling and Molecular Docking were conducted to filter the optimal candidate as the subsequent adjuvant or alternative for IFX in predicted non-responders. At last, the down-regulation of the key model-gene/protein CYP24A1 by the drug candidate Deferasirox was verified by Western Blot and qRT-PCR Assay based on cellular experiments. RESULTS A total of 113 DEGs were identified in the Training Cohort, mainly enriched in inflammatory cell chemotaxis, migration, and response to molecules derived from intestinal microbiota. Activated pro-inflammatory innate immune cells, including neutrophils, M1 macrophages, activated dendritic cells and mast cells, were significantly enriched in colons of non-responders. The prediction model based on three model-genes (IFI44L, CYP24A1, and RGS1) exhibited strong predictive efficacy, with AUC values of 0.901 and 0.80 in the Training and Validation Cohorts, respectively. Higher expression of the three model-genes/proteins in colons of non-responders to IFX was confirmed by clinical colonic mucosal biopsies. 4 Drugs (Calcitriol, Lunacalcipol, Deferasirox, Telaprevir), 15 miRNAs and 66 corresponding lnRNAs interacting with model-genes were identified. The protein 3D structure of the key model-gene/protein (human-derived CYP24A1) was developed. Through the Molecular Docking and cellular experimental validation, Deferasirox, which significantly down-regulated both the RNA and protein expression of CYP24A1, was identified as the optimal adjuvant or alternative for IFX in predicted non-responders with UC. CONCLUSION This study developed a novel prediction model for pre-assessing the efficacy of IFX in patients with UC, as the first step towards personalized therapy. Meanwhile, drugs and non-coding RNAs were provided as potential candidates to develop the next-step precise treatment for the predicted non-responders. In particular, Defeasirox appears to hold promise as an adjuvant or alternative to IFX for the optimization of UC therapy.
Collapse
Affiliation(s)
- Dan Pu
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Pengfei Wang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Xiang Wang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Yonggang Tian
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Hang Gong
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Xueni Ma
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Muyang Li
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Dekui Zhang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China; Key Laboratory of Digestive Diseases, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou City, Gansu Province, China.
| |
Collapse
|
2
|
Salisbury SJ, Daniels RR, Monaghan SJ, Bron JE, Villamayor PR, Gervais O, Fast MD, Sveen L, Houston RD, Robinson N, Robledo D. Keratinocytes drive the epithelial hyperplasia key to sea lice resistance in coho salmon. BMC Biol 2024; 22:160. [PMID: 39075472 PMCID: PMC11287951 DOI: 10.1186/s12915-024-01952-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 06/28/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Salmonid species have followed markedly divergent evolutionary trajectories in their interactions with sea lice. While sea lice parasitism poses significant economic, environmental, and animal welfare challenges for Atlantic salmon (Salmo salar) aquaculture, coho salmon (Oncorhynchus kisutch) exhibit near-complete resistance to sea lice, achieved through a potent epithelial hyperplasia response leading to rapid louse detachment. The molecular mechanisms underlying these divergent responses to sea lice are unknown. RESULTS We characterized the cellular and molecular responses of Atlantic salmon and coho salmon to sea lice using single-nuclei RNA sequencing. Juvenile fish were exposed to copepodid sea lice (Lepeophtheirus salmonis), and lice-attached pelvic fin and skin samples were collected 12 h, 24 h, 36 h, 48 h, and 60 h after exposure, along with control samples. Comparative analysis of control and treatment samples revealed an immune and wound-healing response that was common to both species, but attenuated in Atlantic salmon, potentially reflecting greater sea louse immunomodulation. Our results revealed unique but complementary roles of three layers of keratinocytes in the epithelial hyperplasia response leading to rapid sea lice rejection in coho salmon. Our results suggest that basal keratinocytes direct the expansion and mobility of intermediate and, especially, superficial keratinocytes, which eventually encapsulate the parasite. CONCLUSIONS Our results highlight the key role of keratinocytes in coho salmon's sea lice resistance and the diverged biological response of the two salmonid host species when interacting with this parasite. This study has identified key pathways and candidate genes that could be manipulated using various biotechnological solutions to improve Atlantic salmon sea lice resistance.
Collapse
Affiliation(s)
- S J Salisbury
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
| | - R Ruiz Daniels
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - S J Monaghan
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | - J E Bron
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | - P R Villamayor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
- Department of Genetics, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - O Gervais
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - M D Fast
- Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | | | - R D Houston
- Benchmark Genetics, 1 Pioneer BuildingMilton Bridge, Edinburgh TechnopolePenicuik, UK
| | - N Robinson
- Nofima AS, Tromsø, Norway.
- Sustainable Aquaculture Laboratory - Temperate and Tropical (SALTT), Deakin University, Melbourne, VIC, 3225, Australia.
| | - D Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
- Department of Genetics, University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
3
|
Lyu M, Xu G, Zhou J, Reboud J, Wang Y, Lai H, Chen Y, Zhou Y, Zhu G, Cooper JM, Ying B. Single-Cell Sequencing Reveals Functional Alterations in Tuberculosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305592. [PMID: 38192178 PMCID: PMC10953544 DOI: 10.1002/advs.202305592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/21/2023] [Indexed: 01/10/2024]
Abstract
Despite its importance, the functional heterogeneity surrounding the dynamics of interactions between mycobacterium tuberculosis and human immune cells in determining host immune strength and tuberculosis (TB) outcomes, remains far from understood. This work now describes the development of a new technological platform to elucidate the immune function differences in individuals with TB, integrating single-cell RNA sequencing and cell surface antibody sequencing to provide both genomic and phenotypic information from the same samples. Single-cell analysis of 23 990 peripheral blood mononuclear cells from a new cohort of primary TB patients and healthy controls enables to not only show four distinct immune phenotypes (TB, myeloid, and natural killer (NK) cells), but also determine the dynamic changes in cell population abundance, gene expression, developmental trajectory, transcriptomic regulation, and cell-cell signaling. In doing so, TB-related changes in immune cell functions demonstrate that the immune response is mediated through host T cells, myeloid cells, and NK cells, with TB patients showing decreased naive, cytotoxicity, and memory functions of T cells, rather than their immunoregulatory function. The platform also has the potential to identify new targets for immunotherapeutic treatment strategies to restore T cells from dysfunctional or exhausted states.
Collapse
Affiliation(s)
- Mengyuan Lyu
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Gaolian Xu
- School of Biomedical Engineering/Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Jian Zhou
- Department of Thoracic SurgeryWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Julien Reboud
- Division of Biomedical EngineeringUniversity of GlasgowGlasgowG12 8LTUnited Kingdom
| | - Yili Wang
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Hongli Lai
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Yi Chen
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Yanbing Zhou
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Guiying Zhu
- School of Biomedical Engineering/Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Jonathan M. Cooper
- Division of Biomedical EngineeringUniversity of GlasgowGlasgowG12 8LTUnited Kingdom
| | - Binwu Ying
- Department of Laboratory MedicineWest China HospitalSichuan UniversityChengduSichuan610041P. R. China
| |
Collapse
|
4
|
Luo VM, Shen C, Worme S, Bhagrath A, Simo-Cheyou E, Findlay S, Hébert S, Wai Lam Poon W, Aryanpour Z, Zhang T, Zahedi RP, Boulais J, Buchwald ZS, Borchers CH, Côté JF, Kleinman CL, Mandl JN, Orthwein A. The Deubiquitylase Otub1 Regulates the Chemotactic Response of Splenic B Cells by Modulating the Stability of the γ-Subunit Gng2. Mol Cell Biol 2024; 44:1-16. [PMID: 38270191 PMCID: PMC10829841 DOI: 10.1080/10985549.2023.2290434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/28/2023] [Indexed: 01/26/2024] Open
Abstract
The ubiquitin proteasome system performs the covalent attachment of lysine 48-linked polyubiquitin chains to substrate proteins, thereby targeting them for degradation, while deubiquitylating enzymes (DUBs) reverse this process. This posttranslational modification regulates key features both of innate and adaptative immunity, including antigen presentation, protein homeostasis and signal transduction. Here we show that loss of one of the most highly expressed DUBs, Otub1, results in changes in murine splenic B cell subsets, leading to a significant increase in marginal zone and transitional B cells and a concomitant decrease in follicular B cells. We demonstrate that Otub1 interacts with the γ-subunit of the heterotrimeric G protein, Gng2, and modulates its ubiquitylation status, thereby controlling Gng2 stability. Proximal mapping of Gng2 revealed an enrichment in partners associated with chemokine signaling, actin cytoskeleton and cell migration. In line with these findings, we show that Otub1-deficient B cells exhibit greater Ca2+ mobilization, F-actin polymerization and chemotactic responsiveness to Cxcl12, Cxcl13 and S1P in vitro, which manifests in vivo as altered localization of B cells within the spleen. Together, our data establishes Otub1 as a novel regulator of G-protein coupled receptor signaling in B cells, regulating their differentiation and positioning in the spleen.
Collapse
Affiliation(s)
- Vincent M. Luo
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - Connie Shen
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- McGill Research Centre for Complex Traits, McGill University, Montréal, Québec, Canada
| | - Samantha Worme
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | - Aanya Bhagrath
- McGill Research Centre for Complex Traits, McGill University, Montréal, Québec, Canada
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - Estelle Simo-Cheyou
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Steven Findlay
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | - Steven Hébert
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
| | - William Wai Lam Poon
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | - Zahra Aryanpour
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - Thomas Zhang
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - René P. Zahedi
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- CancerCare Manitoba Research Institute, Winnipeg, Manitoba, Canada
| | - Jonathan Boulais
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Québec, Canada
| | - Zachary S. Buchwald
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Christoph H. Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
- Gerald Bronfman Department of Oncology, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Department of Pathology, McGill University, Montreal, Québec, Canada
| | - Jean-Francois Côté
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Québec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montreal, Québec, Canada
- Département de Médecine (Programmes de Biologie Moléculaire), Université de Montréal, Montreal, Québec, Canada
| | - Claudia L. Kleinman
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
| | - Judith N. Mandl
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- McGill Research Centre for Complex Traits, McGill University, Montréal, Québec, Canada
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - Alexandre Orthwein
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
- Gerald Bronfman Department of Oncology, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| |
Collapse
|
5
|
Flynn AL, Gans J, Escobedo J, Zhu C, Florescu AM, Shankara S, Madden SL, Kim PS, Pao LI. RGS1 Modulates Autophagic and Metabolic Programs and Is a Critical Mediator of Human Regulatory T Cell Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1656-1668. [PMID: 37850953 DOI: 10.4049/jimmunol.2200402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/27/2023] [Indexed: 10/19/2023]
Abstract
Regulatory T cells (Tregs) are critical mediators of immune tolerance and play a diametric role in cancer and autoimmunity. Tumor-infiltrating Tregs are often associated with poor prognosis in solid tumors because their enrichment in the tumor microenvironment contributes to immunosuppression. Conversely, dysregulation in the Treg compartment can disrupt self-tolerance, leading to autoimmunity. In the present study, we describe what is, to our knowledge, a novel regulator of Tregs, the GTPase activator regulator of G protein 1 (RGS1), demonstrating that RGS1-deficient human Tregs show downregulation of Treg-associated genes and are less immunosuppressive. These RGS1-deficient Tregs exhibit perturbations to the FOXP3-c-MYC transcriptional axis and downstream metabolic and autophagy programs by shifting their energy demands toward glycolysis and rendering them less autophagic. Taken together, RGS1 may serve as an apical node of Treg function by regulating the FOXP3-c-MYC transcriptional axis, thereby providing a therapeutic rationale for targeting RGS1 for treatment of cancer and autoimmune diseases.
Collapse
Affiliation(s)
| | - Joseph Gans
- Department of Translational Sciences, Sanofi, Cambridge, MA
| | | | - Cheng Zhu
- Department of Translational Sciences, Sanofi, Cambridge, MA
| | | | | | | | - Peter S Kim
- Department of Oncology, Sanofi, Cambridge, MA
| | - Lily I Pao
- Department of Oncology, Sanofi, Cambridge, MA
| |
Collapse
|
6
|
Yazicioglu YF, Marin E, Sandhu C, Galiani S, Raza IGA, Ali M, Kronsteiner B, Compeer EB, Attar M, Dunachie SJ, Dustin ML, Clarke AJ. Dynamic mitochondrial transcription and translation in B cells control germinal center entry and lymphomagenesis. Nat Immunol 2023; 24:991-1006. [PMID: 37095377 PMCID: PMC10232359 DOI: 10.1038/s41590-023-01484-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 03/09/2023] [Indexed: 04/26/2023]
Abstract
Germinal center (GC) B cells undergo proliferation at very high rates in a hypoxic microenvironment but the cellular processes driving this are incompletely understood. Here we show that the mitochondria of GC B cells are highly dynamic, with significantly upregulated transcription and translation rates associated with the activity of transcription factor A, mitochondrial (TFAM). TFAM, while also necessary for normal B cell development, is required for entry of activated GC precursor B cells into the germinal center reaction; deletion of Tfam significantly impairs GC formation, function and output. Loss of TFAM in B cells compromises the actin cytoskeleton and impairs cellular motility of GC B cells in response to chemokine signaling, leading to their spatial disorganization. We show that B cell lymphoma substantially increases mitochondrial translation and that deletion of Tfam in B cells is protective against the development of lymphoma in a c-Myc transgenic mouse model. Finally, we show that pharmacological inhibition of mitochondrial transcription and translation inhibits growth of GC-derived human lymphoma cells and induces similar defects in the actin cytoskeleton.
Collapse
Affiliation(s)
| | - Eros Marin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Ciaran Sandhu
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Medical Sciences Division, University of Oxford, Oxford, UK
| | - Silvia Galiani
- Medical Research Centre Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Iwan G A Raza
- Medical Sciences Division, University of Oxford, Oxford, UK
| | - Mohammad Ali
- Nuffield Department of Medicine Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Barbara Kronsteiner
- Nuffield Department of Medicine Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Ewoud B Compeer
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Moustafa Attar
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Susanna J Dunachie
- Nuffield Department of Medicine Centre For Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- National Institute for Health and Care Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | |
Collapse
|
7
|
von Werdt D, Gungor B, Barreto de Albuquerque J, Gruber T, Zysset D, Kwong Chung CKC, Corrêa-Ferreira A, Berchtold R, Page N, Schenk M, Kehrl JH, Merkler D, Imhof BA, Stein JV, Abe J, Turchinovich G, Finke D, Hayday AC, Corazza N, Mueller C. Regulator of G-protein signaling 1 critically supports CD8 + T RM cell-mediated intestinal immunity. Front Immunol 2023; 14:1085895. [PMID: 37153600 PMCID: PMC10158727 DOI: 10.3389/fimmu.2023.1085895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/13/2023] [Indexed: 05/09/2023] Open
Abstract
Members of the Regulator of G-protein signaling (Rgs) family regulate the extent and timing of G protein signaling by increasing the GTPase activity of Gα protein subunits. The Rgs family member Rgs1 is one of the most up-regulated genes in tissue-resident memory (TRM) T cells when compared to their circulating T cell counterparts. Functionally, Rgs1 preferentially deactivates Gαq, and Gαi protein subunits and can therefore also attenuate chemokine receptor-mediated immune cell trafficking. The impact of Rgs1 expression on tissue-resident T cell generation, their maintenance, and the immunosurveillance of barrier tissues, however, is only incompletely understood. Here we report that Rgs1 expression is readily induced in naïve OT-I T cells in vivo following intestinal infection with Listeria monocytogenes-OVA. In bone marrow chimeras, Rgs1 -/- and Rgs1 +/+ T cells were generally present in comparable frequencies in distinct T cell subsets of the intestinal mucosa, mesenteric lymph nodes, and spleen. After intestinal infection with Listeria monocytogenes-OVA, however, OT-I Rgs1 +/+ T cells outnumbered the co-transferred OT-I Rgs1- /- T cells in the small intestinal mucosa already early after infection. The underrepresentation of the OT-I Rgs1 -/- T cells persisted to become even more pronounced during the memory phase (d30 post-infection). Remarkably, upon intestinal reinfection, mice with intestinal OT-I Rgs1 +/+ TRM cells were able to prevent the systemic dissemination of the pathogen more efficiently than those with OT-I Rgs1 -/- TRM cells. While the underlying mechanisms are not fully elucidated yet, these data thus identify Rgs1 as a critical regulator for the generation and maintenance of tissue-resident CD8+ T cells as a prerequisite for efficient local immunosurveillance in barrier tissues in case of reinfections with potential pathogens.
Collapse
Affiliation(s)
- Diego von Werdt
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Bilgi Gungor
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Thomas Gruber
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Daniel Zysset
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Cheong K. C. Kwong Chung
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Gastrointestinal Health, Immunology, Nestlé Research, Lausanne, Switzerland
| | - Antonia Corrêa-Ferreira
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Regina Berchtold
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Nicolas Page
- Department of Pathology, Division of Clinical Pathology, University & University Hospitals of Geneva, Geneva, Switzerland
| | - Mirjam Schenk
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - John H. Kehrl
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Doron Merkler
- Department of Pathology, Division of Clinical Pathology, University & University Hospitals of Geneva, Geneva, Switzerland
| | - Beat A. Imhof
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Pathology and Immunology, Centre Medical Universitaire, University of Geneva, Geneva, Switzerland
| | - Jens V. Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Gleb Turchinovich
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Finke
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Adrian C. Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Nadia Corazza
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- *Correspondence: Christoph Mueller, ; Nadia Corazza,
| | - Christoph Mueller
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
- *Correspondence: Christoph Mueller, ; Nadia Corazza,
| |
Collapse
|
8
|
Xue W, He W, Yan M, Zhao H, Pi J. Exploring Shared Biomarkers of Myocardial Infarction and Alzheimer's Disease via Single-Cell/Nucleus Sequencing and Bioinformatics Analysis. J Alzheimers Dis 2023; 96:705-723. [PMID: 37840493 PMCID: PMC10657707 DOI: 10.3233/jad-230559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Patients are at increased risk of dementia, including Alzheimer's disease (AD), after myocardial infarction (MI), but the biological link between MI and AD is unclear. OBJECTIVE To understand the association between the pathogenesis of MI and AD and identify common biomarkers of both diseases. METHODS Using public databases, we identified common biomarkers of MI and AD. Least absolute shrinkage and selection operator (LASSO) regression and protein-protein interaction (PPI) network were performed to further screen hub biomarkers. Functional enrichment analyses were performed on the hub biomarkers. Single-cell/nucleus analysis was utilized to further analyze the hub biomarkers at the cellular level in carotid atherosclerosis and AD datasets. Motif enrichment analysis was used to screen key transcription factors. RESULTS 26 common differentially expressed genes were screened between MI and AD. Function enrichment analyses showed that these differentially expressed genes were mainly associated with inflammatory pathways. A key gene, Regulator of G-protein Signaling 1 (RGS1), was obtained by LASSO regression and PPI network. RGS1 was confirmed to mainly express in macrophages and microglia according to single-cell/nucleus analysis. The difference in expression of RGS1 in macrophages and microglia between disease groups and controls was statistically significant (p < 0.0001). The expression of RGS1 in the disease groups was upregulated with the differentiation of macrophages and microglia. RelA was a key transcription factor regulating RGS1. CONCLUSION Macrophages and microglia are involved in the inflammatory response of MI and AD. RGS1 may be a key biomarker in this process.
Collapse
Affiliation(s)
- Weiqi Xue
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weifeng He
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mengyuan Yan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huanyi Zhao
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianbin Pi
- Department of Cardiovascular Disease, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| |
Collapse
|
9
|
Álvarez L, Marín-García PJ, Llobat L. Immunological and genomic characterization of Ibizan Hound dogs in an endemic Leishmania infantum region. Parasit Vectors 2022; 15:445. [DOI: 10.1186/s13071-022-05504-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Abstract
Background
The Ibizan Hound is a canine breed native to the Mediterranean region, where leishmaniosis is an endemic zoonosis. Several studies indicate low prevalence of this disease in these dogs but the underlying molecular mechanism remains unknown.
Methods
In this study, qualitative immunological and genomic profiles of this breed have been analyzed.
Results
Our analysis shows relevant differences between the cytokine serum profile of Ibizan Hound dogs and previously published data from other canine strains. Additionally, several genetic risk variants related to the immune response, regulation of the immune system, and genes encoding cytokines and their receptors have been studied. The most relevant genes that presented such fixed polymorphisms were IFNG and IL6R. Other variants with frequencies ≥ 0.7 were found in the genes ARHGAP18, DAPK1, GNAI2, MITF, IL12RB1, LTBP1, SCL28A3, SCL35D2, PTPN22, CIITA, THEMIS, and CD180. Epigenetic regulatory genes such as HEY2 and L3MBTL3 showed also intronic polymorphisms.
Conclusions
Our analysis and results indicate that the regulation of immune responses is different in Ibizan Hounds compared to other breeds. Future studies are needed to elucidate whether these differences are related to the low prevalence of L. infantum infection in the Ibizan Hound.
Graphical Abstract
Collapse
|
10
|
Rodríguez-Fernández JL, Criado-García O. A meta-analysis indicates that the regulation of cell motility is a non-intrinsic function of chemoattractant receptors that is governed independently of directional sensing. Front Immunol 2022; 13:1001086. [PMID: 36341452 PMCID: PMC9630654 DOI: 10.3389/fimmu.2022.1001086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
Chemoattraction, defined as the migration of a cell toward a source of a chemical gradient, is controlled by chemoattractant receptors. Chemoattraction involves two basic activities, namely, directional sensing, a molecular mechanism that detects the direction of a source of chemoattractant, and actin-based motility, which allows the migration of a cell towards it. Current models assume first, that chemoattractant receptors govern both directional sensing and motility (most commonly inducing an increase in the migratory speed of the cells, i.e. chemokinesis), and, second, that the signaling pathways controlling both activities are intertwined. We performed a meta-analysis to reassess these two points. From this study emerge two main findings. First, although many chemoattractant receptors govern directional sensing, there are also receptors that do not regulate cell motility, suggesting that is the ability to control directional sensing, not motility, that best defines a chemoattractant receptor. Second, multiple experimental data suggest that receptor-controlled directional sensing and motility can be controlled independently. We hypothesize that this independence may be based on the existence of separated signalling modules that selectively govern directional sensing and motility in chemotactic cells. Together, the information gathered can be useful to update current models representing the signalling from chemoattractant receptors. The new models may facilitate the development of strategies for a more effective pharmacological modulation of chemoattractant receptor-controlled chemoattraction in health and disease.
Collapse
|
11
|
Lu X, Zhang K, Jiang W, Li H, Huang Y, Du M, Wan J, Cao Y, Du L, Liu X, Pan W. Single-cell RNA sequencing combined with whole exome sequencing reveals the landscape of the immune pathogenic response to chronic mucocutaneous candidiasis with STAT1 GOF mutation. Front Immunol 2022; 13:988766. [PMID: 36225936 PMCID: PMC9549386 DOI: 10.3389/fimmu.2022.988766] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic mucocutaneous candidiasis (CMC) is characterized by recurrent or persistent infections with Candida of the skin, nails, and mucous membranes (e.g., mouth, esophagus, and vagina). Compared with that of other infectious diseases, the immune pathogenic mechanism of CMC is still poorly understood. We identified a signal transducer and activator of transcription 1 gain-of-function (c.Y289C) mutation in a CMC patient. Single-cell transcriptional profiling on peripheral blood mononuclear cells from this patient revealed decreases in immature B cells and monocytes. Further analysis revealed several differentially expressed genes related to immune regulation, including RGS1, TNFAIP3, S100A8/A9, and CTSS. In our review of the literature on signal transducer and activator of transcription 1 gain-of-function (c.Y289C) mutations, we identified seven cases in total. The median age of onset for CMC (n=4, data lacking for three cases) was 10.5 years (range: birth to 11 years), with an average onset age of 8 years. There were no reports linking tumors to the c.Y289C mutation, and the incidence of pre-existing clinical disease in patients with the c.Y289C mutation was similar to previous data.
Collapse
Affiliation(s)
- Xiaodi Lu
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Keming Zhang
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Weiwei Jiang
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
- Department of Dermatology, 72nd Group Army Hospital of People’s Liberation Army (PLA), Huzhou, China
| | - Hang Li
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Yue Huang
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Mingwei Du
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Jian Wan
- Department of Dermatology, Pudong New Area People’s Hospital, Shanghai, China
| | - Yanyun Cao
- Department of Dermatology, Pudong New Area People’s Hospital, Shanghai, China
- Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Pudong New Area People’s Hospital, Shanghai, China
| | - Lin Du
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
- *Correspondence: Lin Du, ; Xiaogang Liu, ; Weihua Pan,
| | - Xiaogang Liu
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
- *Correspondence: Lin Du, ; Xiaogang Liu, ; Weihua Pan,
| | - Weihua Pan
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
- *Correspondence: Lin Du, ; Xiaogang Liu, ; Weihua Pan,
| |
Collapse
|
12
|
Wang B, Wang M, Ao D, Wei X. CXCL13-CXCR5 axis: Regulation in inflammatory diseases and cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188799. [PMID: 36103908 DOI: 10.1016/j.bbcan.2022.188799] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 01/10/2023]
Abstract
Chemokine C-X-C motif ligand 13 (CXCL13), originally identified as a B-cell chemokine, plays an important role in the immune system. The interaction between CXCL13 and its receptor, the G-protein coupled receptor (GPCR) CXCR5, builds a signaling network that regulates not only normal organisms but also the development of many diseases. However, the precise action mechanism remains unclear. In this review, we discussed the functional mechanisms of the CXCL13-CXCR5 axis under normal conditions, with special focus on its association with diseases. For certain refractory diseases, we emphasize the diagnostic and therapeutic role of CXCL13-CXCR5 axis.
Collapse
Affiliation(s)
- Binhan Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Danyi Ao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Villaseca S, Romero G, Ruiz MJ, Pérez C, Leal JI, Tovar LM, Torrejón M. Gαi protein subunit: A step toward understanding its non-canonical mechanisms. Front Cell Dev Biol 2022; 10:941870. [PMID: 36092739 PMCID: PMC9449497 DOI: 10.3389/fcell.2022.941870] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
The heterotrimeric G protein family plays essential roles during a varied array of cellular events; thus, its deregulation can seriously alter signaling events and the overall state of the cell. Heterotrimeric G-proteins have three subunits (α, β, γ) and are subdivided into four families, Gαi, Gα12/13, Gαq, and Gαs. These proteins cycle between an inactive Gα-GDP state and active Gα-GTP state, triggered canonically by the G-protein coupled receptor (GPCR) and by other accessory proteins receptors independent also known as AGS (Activators of G-protein Signaling). In this review, we summarize research data specific for the Gαi family. This family has the largest number of individual members, including Gαi1, Gαi2, Gαi3, Gαo, Gαt, Gαg, and Gαz, and constitutes the majority of G proteins α subunits expressed in a tissue or cell. Gαi was initially described by its inhibitory function on adenylyl cyclase activity, decreasing cAMP levels. Interestingly, today Gi family G-protein have been reported to be importantly involved in the immune system function. Here, we discuss the impact of Gαi on non-canonical effector proteins, such as c-Src, ERK1/2, phospholipase-C (PLC), and proteins from the Rho GTPase family members, all of them essential signaling pathways regulating a wide range of physiological processes.
Collapse
|
14
|
Chan KYY, Chung PY, Zhang C, Poon ENY, Leung AWK, Leung KT. R4 RGS proteins as fine tuners of immature and mature hematopoietic cell trafficking. J Leukoc Biol 2022; 112:785-797. [PMID: 35694792 DOI: 10.1002/jlb.1mr0422-475r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are the largest and most diverse group of membrane receptors. They are involved in almost every physiologic process and consequently have a pivotal role in an extensive number of pathologies, including genetic, neurologic, and immune system disorders. Indeed, the vast array of GPCRs mechanisms have led to the development of a tremendous number of drug therapies and already account for about a third of marketed drugs. These receptors mediate their downstream signals primarily via G proteins. The regulators of G-protein signaling (RGS) proteins are now in the spotlight as the critical modulatory factors of active GTP-bound Gα subunits of heterotrimeric G proteins to fine-tune the biologic responses driven by the GPCRs. Also, they possess noncanonical functions by multiple mechanisms, such as protein-protein interactions. Essential roles and impacts of these RGS proteins have been revealed in physiology, including hematopoiesis and immunity, and pathologies, including asthma, cancers, and neurologic disorders. This review focuses on the largest subfamily of R4 RGS proteins and provides a brief overview of their structures and G-proteins selectivity. With particular interest, we explore and highlight, their expression in the hematopoietic system and the regulation in the engraftment of hematopoietic stem/progenitor cells (HSPCs). Distinct expression patterns of R4 RGS proteins in the hematopoietic system and their pivotal roles in stem cell trafficking pave the way for realizing new strategies for enhancing the clinical performance of hematopoietic stem cell transplantation. Finally, we discuss the exciting future trends in drug development by targeting RGS activity and expression with small molecules inhibitors and miRNA approaches.
Collapse
Affiliation(s)
- Kathy Yuen Yee Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Po Yee Chung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chi Zhang
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ellen Ngar Yun Poon
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Alex Wing Kwan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Department of Paediatrics & Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong SAR, China
| | - Kam Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
15
|
Shen M, Li T, Feng Y, Chen Z, Dou T, Wu P, Wang K, Lu J, Qu L. Exploring the expression and preliminary function of chicken regulator of G protein signalling 3 ( RGS3) gene in follicular development. Br Poult Sci 2022; 63:613-620. [PMID: 35522181 DOI: 10.1080/00071668.2022.2071597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
1. The following study explored the expression and preliminary function of RGS3. The spatial and temporal expression patterns of the RGS3 gene were analysed in the ovarian stroma of Shendan No. 6 Green shell hens and Hy-line Brown hens at four time points (6, 28, 40 and 52 weeks old), as well as in various organs and follicles of Hy-line Brown hens.2. Based on the genomic and protein sequences of RGS3 in NCBI database, phylogenetic trees were constructed using MEGA-X. The protein interaction network was analysed using STRING. According to the results of protein-protein interaction network and pathways, the mRNA expression levels of RGS3 and three interaction proteins were explored by qRT-PCR in vitro.3. Spatio-temporal expression data revealed that RGS3 mRNA was expressed in all the organs tested, being highest in the hypothalamus. In different follicles, RGS3 mRNA was highly expressed in post-ovulatory follicles, followed by ovarian stroma and large white follicles. The expression levels of RGS3 mRNA in the ovarian stroma were significantly higher in Shendan No. 6 Green shell hens than that in the Hy-line Brown hens at all egg-laying stages.4. The phylogenetic tree results showed that ducks, geese and chickens had higher homology based on the genomic and protein sequence of RGS3. Moreover, chicken RGS3 interacted with GSK3B, RAF1 and BRAF based on STRING prediction. In vitro follicle stimulating hormone (FSH) treatment showed that mRNA expression levels of RGS3 and those of its predicted interacting proteins BRAF and GSK3B decreased with increasing FSH concentration. The results suggested that RGS3 responds to FSH and may play an important role in the regulation follicular development in chicken.
Collapse
Affiliation(s)
- Manman Shen
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 225108, China.,Jiangsu Institute of Poultry Science, Chinese Academy of Agricultural Sciences, Yangzhou, 225125, China.,Jiangsu Key Laboratory of Animal genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Tao Li
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 225108, China
| | - Yuan Feng
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 225108, China
| | - Zikang Chen
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 225108, China
| | - Taocun Dou
- Jiangsu Institute of Poultry Science, Chinese Academy of Agricultural Sciences, Yangzhou, 225125, China
| | - Ping Wu
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 225108, China
| | - Kehua Wang
- Jiangsu Institute of Poultry Science, Chinese Academy of Agricultural Sciences, Yangzhou, 225125, China
| | - Jian Lu
- Jiangsu Institute of Poultry Science, Chinese Academy of Agricultural Sciences, Yangzhou, 225125, China
| | - Liang Qu
- Jiangsu Institute of Poultry Science, Chinese Academy of Agricultural Sciences, Yangzhou, 225125, China
| |
Collapse
|
16
|
Sun L, Shen F, Xiong Z, Yang H, Dong Z, Xiang J, Gu Q, Ji Q, Fan C, Liu Z. DNA Engineered Lymphocyte-Based Homologous Targeting Artificial Antigen-Presenting Cells for Personalized Cancer Immunotherapy. J Am Chem Soc 2022; 144:7634-7645. [PMID: 35438987 DOI: 10.1021/jacs.1c09316] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Artificial antigen-presenting cells (aAPCs) constructed by integrating T cell activation ligands on biocompatible materials hold great potential in tumor immunotherapy. However, it remains challenging to develop aAPCs, which could mimic the characteristics of natural APCs, thereby realizing antigen-specific T cells activation in vivo. Here, we report the first effort to construct natural lymphocyte-based homologous targeting aAPCs (LC-aAPCs) with lipid-DNA-mediated noninvasive live cell surface engineering. Through a predesigned bottom-up self-assembly path, we achieved natural-APC-mimicking distribution of T cell activation ligands on LC-aAPCs, which would enable the optimized T cell activation. Moreover, the lipid-DNA-mediated self-assembly occurring on lipid bilayers would not affect the functions of homing receptors expressed on lymphocyte. Therefore, such LC-aAPCs could actively migrate to peripheral lymphatic organs and then effectively activate antigen-specific T cells. Combined with an immune checkpoint inhibitor, such LC-aAPCs could effectively inhibit the growth of different tumor models. Thus, our work provides a new design of aAPCs for in vivo applications in tumor immunotherapy, and the lipid-DNA-mediated noninvasive live cell surface engineering would be a powerful tool for designing cell-based therapeutics.
Collapse
Affiliation(s)
- Lele Sun
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Fengyun Shen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Zijian Xiong
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - He Yang
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Ziliang Dong
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jian Xiang
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Qingyang Gu
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Qunsheng Ji
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Chunhai Fan
- Natl Ctr Translat Med, Frontiers Sci Ctr Transformat Mol, Sch Chem & Chem Engn, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
17
|
Neuron-derived neuropeptide Y fine-tunes the splenic immune responses. Neuron 2022; 110:1327-1339.e6. [PMID: 35139365 DOI: 10.1016/j.neuron.2022.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/14/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022]
Abstract
The nervous and immune systems are closely entwined to maintain the immune balance in health and disease. Here, we showed that LPS can activate suprarenal and celiac ganglia (SrG-CG) neurons and upregulate NPY expression in rats. Single-cell sequencing analysis revealed that knockdown of the NPY gene in SrG-CG altered the proliferation and activation of splenic lymphocytes. In a neuron and splenocyte coculture system and in vivo experiments, neuronal NPY in SrG-CG attenuated the splenic immune response. Notably, we demonstrated that neuronal NPF in Drosophila exerted a conservative immunomodulatory effect. Moreover, numerous SNPs in NPY and its receptors were significantly associated with human autoimmune diseases, which was further supported by the autoimmune disease patients and mouse model experiments. Together, we demonstrated that NPY is an ancient language for nervous-immune system crosstalk and might be utilized to alleviate inflammatory storms during infection and to modulate immune balance in autoimmune diseases.
Collapse
|
18
|
Potential Role of CXCL13/CXCR5 Signaling in Immune Checkpoint Inhibitor Treatment in Cancer. Cancers (Basel) 2022; 14:cancers14020294. [PMID: 35053457 PMCID: PMC8774093 DOI: 10.3390/cancers14020294] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Immunotherapy is currently the backbone of new drug treatments for many cancer patients. CXC chemokine ligand 13 (CXCL13) is an important factor involved in recruiting immune cells that express CXC chemokine receptor type 5 (CXCR5) in the tumor microenvironment and serves as a key molecular determinant of tertiary lymphoid structure (TLS) formation. An increasing number of studies have identified the influence of CXCL13 on prognosis in patients with cancer, regardless of the use of immunotherapy treatment. However, no comprehensive reviews of the role of CXCL13 in cancer immunotherapy have been published to date. This review aims to provide an overview of the CXCL13/CXCR5 signaling axis to summarize its mechanisms of action in cancer cells and lymphocytes, in addition to effects on immunity and cancer pathobiology, and its potential as a biomarker for the response to cancer immunotherapy. Abstract Immune checkpoint inhibitors (ICIs), including antibodies that target programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), or cytotoxic T lymphocyte antigen 4 (CTLA4), represent some of the most important breakthroughs in new drug development for oncology therapy from the past decade. CXC chemokine ligand 13 (CXCL13) exclusively binds CXC chemokine receptor type 5 (CXCR5), which plays a critical role in immune cell recruitment and activation and the regulation of the adaptive immune response. CXCL13 is a key molecular determinant of the formation of tertiary lymphoid structures (TLSs), which are organized aggregates of T, B, and dendritic cells that participate in the adaptive antitumor immune response. CXCL13 may also serve as a prognostic and predictive factor, and the role played by CXCL13 in some ICI-responsive tumor types has gained intense interest. This review discusses how CXCL13/CXCR5 signaling modulates cancer and immune cells to promote lymphocyte infiltration, activation by tumor antigens, and differentiation to increase the antitumor immune response. We also summarize recent preclinical and clinical evidence regarding the ICI-therapeutic implications of targeting the CXCL13/CXCR5 axis and discuss the potential role of this signaling pathway in cancer immunotherapy.
Collapse
|
19
|
Frattolin J, Watson DJ, Bonneuil WV, Russell MJ, Fasanella Masci F, Bandara M, Brook BS, Nibbs RJB, Moore JE. The Critical Importance of Spatial and Temporal Scales in Designing and Interpreting Immune Cell Migration Assays. Cells 2021; 10:3439. [PMID: 34943947 PMCID: PMC8700135 DOI: 10.3390/cells10123439] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023] Open
Abstract
Intravital microscopy and other direct-imaging techniques have allowed for a characterisation of leukocyte migration that has revolutionised the field of immunology, resulting in an unprecedented understanding of the mechanisms of immune response and adaptive immunity. However, there is an assumption within the field that modern imaging techniques permit imaging parameters where the resulting cell track accurately captures a cell's motion. This notion is almost entirely untested, and the relationship between what could be observed at a given scale and the underlying cell behaviour is undefined. Insufficient spatial and temporal resolutions within migration assays can result in misrepresentation of important physiologic processes or cause subtle changes in critical cell behaviour to be missed. In this review, we contextualise how scale can affect the perceived migratory behaviour of cells, summarise the limited approaches to mitigate this effect, and establish the need for a widely implemented framework to account for scale and correct observations of cell motion. We then extend the concept of scale to new approaches that seek to bridge the current "black box" between single-cell behaviour and systemic response.
Collapse
Affiliation(s)
- Jennifer Frattolin
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Daniel J. Watson
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Willy V. Bonneuil
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Matthew J. Russell
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (M.J.R.); (B.S.B.)
| | - Francesca Fasanella Masci
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - Mikaila Bandara
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - Bindi S. Brook
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (M.J.R.); (B.S.B.)
| | - Robert J. B. Nibbs
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - James E. Moore
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| |
Collapse
|
20
|
R4 RGS proteins suppress engraftment of human hematopoietic stem/progenitor cells by modulating SDF-1/CXCR4 signaling. Blood Adv 2021; 5:4380-4392. [PMID: 34500454 PMCID: PMC8579266 DOI: 10.1182/bloodadvances.2020003307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
Specific R4 RGS members are expressed in human HSPCs and regulated by the SDF-1/CXCR4 axis. RGS1/13/16 suppress HSPC engraftment, SDF-1 signaling, and key effectors of stem cell trafficking/maintenance.
Homing and engraftment of hematopoietic stem/progenitor cells (HSPCs) into the bone marrow (BM) microenvironment are tightly regulated by the chemokine stromal cell–derived factor-1 (SDF-1) and its G-protein–coupled receptor C-X-C motif chemokine receptor 4 (CXCR4), which on engagement with G-protein subunits, trigger downstream migratory signals. Regulators of G-protein signaling (RGS) are GTPase-accelerating protein of the Gα subunit and R4 subfamily members have been implicated in SDF-1–directed trafficking of mature hematopoietic cells, yet their expression and influence on HSPCs remain mostly unknown. Here, we demonstrated that human CD34+ cells expressed multiple R4 RGS genes, of which RGS1, RGS2, RGS13, and RGS16 were significantly upregulated by SDF-1 in a CXCR4-dependent fashion. Forced overexpression of RGS1, RGS13, or RGS16 in CD34+ cells not only inhibited SDF-1–directed migration, calcium mobilization, and phosphorylation of AKT, ERK, and STAT3 in vitro, but also markedly reduced BM engraftment in transplanted NOD/SCID mice. Genome-wide microarray analysis of RGS-overexpressing CD34+ cells detected downregulation of multiple effectors with established roles in stem cell trafficking/maintenance. Convincingly, gain-of-function of selected effectors or ex vivo priming with their ligands significantly enhanced HSPC engraftment. We also constructed an evidence-based network illustrating the overlapping mechanisms of RGS1, RGS13, and RGS16 downstream of SDF-1/CXCR4 and Gαi. This model shows that these RGS members mediate compromised kinase signaling and negative regulation of stem cell functions, complement activation, proteolysis, and cell migration. Collectively, this study uncovers an essential inhibitory role of specific R4 RGS proteins in stem cell engraftment, which could potentially be exploited to develop improved clinical HSPC transplantation protocols.
Collapse
|
21
|
Shen Q, Zhang S. Approximate distance correlation for selecting highly interrelated genes across datasets. PLoS Comput Biol 2021; 17:e1009548. [PMID: 34752449 PMCID: PMC8604336 DOI: 10.1371/journal.pcbi.1009548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 11/19/2021] [Accepted: 10/10/2021] [Indexed: 12/13/2022] Open
Abstract
With the rapid accumulation of biological omics datasets, decoding the underlying relationships of cross-dataset genes becomes an important issue. Previous studies have attempted to identify differentially expressed genes across datasets. However, it is hard for them to detect interrelated ones. Moreover, existing correlation-based algorithms can only measure the relationship between genes within a single dataset or two multi-modal datasets from the same samples. It is still unclear how to quantify the strength of association of the same gene across two biological datasets with different samples. To this end, we propose Approximate Distance Correlation (ADC) to select interrelated genes with statistical significance across two different biological datasets. ADC first obtains the k most correlated genes for each target gene as its approximate observations, and then calculates the distance correlation (DC) for the target gene across two datasets. ADC repeats this process for all genes and then performs the Benjamini-Hochberg adjustment to control the false discovery rate. We demonstrate the effectiveness of ADC with simulation data and four real applications to select highly interrelated genes across two datasets. These four applications including 21 cancer RNA-seq datasets of different tissues; six single-cell RNA-seq (scRNA-seq) datasets of mouse hematopoietic cells across six different cell types along the hematopoietic cell lineage; five scRNA-seq datasets of pancreatic islet cells across five different technologies; coupled single-cell ATAC-seq (scATAC-seq) and scRNA-seq data of peripheral blood mononuclear cells (PBMC). Extensive results demonstrate that ADC is a powerful tool to uncover interrelated genes with strong biological implications and is scalable to large-scale datasets. Moreover, the number of such genes can serve as a metric to measure the similarity between two datasets, which could characterize the relative difference of diverse cell types and technologies.
Collapse
Affiliation(s)
- Qunlun Shen
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shihua Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
22
|
Carreras J, Kikuti YY, Hiraiwa S, Miyaoka M, Tomita S, Ikoma H, Ito A, Kondo Y, Itoh J, Roncador G, Martinez A, Colomo L, Hamoudi R, Ando K, Nakamura N. High PTX3 expression is associated with a poor prognosis in diffuse large B-cell lymphoma. Cancer Sci 2021; 113:334-348. [PMID: 34706126 PMCID: PMC8748251 DOI: 10.1111/cas.15179] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/02/2022] Open
Abstract
Tumor‐associated macrophages (TAMs) are associated with a poor prognosis of diffuse large B‐cell lymphoma (DLBCL). As macrophages are heterogeneous, the immune polarization and their pathological role warrant further study. We characterized the microenvironment of DLBCL by immunohistochemistry in a training set of 132 cases, which included 10 Epstein–Barr virus‐encoded small RNA (EBER)‐positive and five high‐grade B‐cell lymphomas, with gene expression profiling in a representative subset of 37 cases. Diffuse large B‐cell lymphoma had a differential infiltration of TAMs. The high infiltration of CD68 (pan‐macrophages), CD16 (M1‐like), CD163, pentraxin 3 (PTX3), and interleukin (IL)‐10‐positive macrophages (M2c‐like) and low infiltration of FOXP3‐positive regulatory T lymphocytes (Tregs) correlated with poor survival. Activated B cell‐like DLBCL was associated with high CD16, CD163, PTX3, and IL‐10, and EBER‐positive DLBCL with high CD163 and PTX3. Programmed cell death‐ligand 1 positively correlated with CD16, CD163, IL‐10, and RGS1. In a multivariate analysis of overall survival, PTX3 and International Prognostic Index were identified as the most relevant variables. The gene expression analysis showed upregulation of genes involved in innate and adaptive immune responses and macrophage and Toll‐like receptor pathways in high PTX3 cases. The prognostic relevance of PTX3 was confirmed in a validation set of 159 cases. Finally, in a series from Europe and North America (GSE10846, R‐CHOP‐like treatment, n = 233) high gene expression of PTX3 correlated with poor survival, and moderately with CSF1R, CD16, MITF, CD163, MYC, and RGS1. Therefore, the high infiltration of M2c‐like immune regulatory macrophages and low infiltration of FOXP3‐positive Tregs is associated with a poor prognosis in DLBCL, for which PTX3 is a new prognostic biomarker.
Collapse
Affiliation(s)
- Joaquim Carreras
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Yara Yukie Kikuti
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Shinichiro Hiraiwa
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Masashi Miyaoka
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Sakura Tomita
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Haruka Ikoma
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Atsushi Ito
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Yusuke Kondo
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Johbu Itoh
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| | - Giovanna Roncador
- Monoclonal Antibodies Core Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | - Antonio Martinez
- Department of Pathology, Hospital Clinic Barcelona, University of Barcelona, Institut d'investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lluis Colomo
- Department of Pathology, Hospital del Mar, Institute Hospital del Mar d'Investigacions Mediques (IMIM), Barcelona, Spain
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Kiyoshi Ando
- Department of Hematology and Oncology, Tokai University, School of Medicine, Isehara, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University, School of Medicine, Isehara, Japan
| |
Collapse
|
23
|
Feng Z, Zhou J, Liu Y, Xia R, Li Q, Yan L, Chen Q, Chen X, Jiang Y, Chao G, Wang M, Zhou G, Zhang Y, Wang Y, Xia H. Epithelium- and endothelium-derived exosomes regulate the alveolar macrophages by targeting RGS1 mediated calcium signaling-dependent immune response. Cell Death Differ 2021; 28:2238-2256. [PMID: 33753901 PMCID: PMC8257848 DOI: 10.1038/s41418-021-00750-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 02/01/2023] Open
Abstract
Alveolar macrophages (AM) maintain airway immune balance; however, the regulation of heterogeneity of AMs is incompletely understood. We demonstrate that RGS1 coregulates the immunophenotype of AM subpopulations, including pro- and anti-inflammatory, injury- and repair-associated, and pro- and antifibrotic phenotypes, through the PLC-IP3R signal-dependent intracellular Ca2+ response. Flt3+ AMs and Tie2+ AMs had different immune properties, and RGS1 expression in the cells was targeted by exosomes (EXOs) containing miR-223 and miR-27b-3p that were derived from vascular endothelial cells (EnCs) and type II alveolar epithelial cells (EpCs-II), respectively. Imbalance of AMs was correlated with acute lung injury/acute respiratory distress syndrome (ALI/ARDS) and pulmonary fibrosis (PF) caused a lack of secretion of CD31+ and CD74+ EXOs derived from EnCs and EpCs-II. Timely treatment with EXOs significantly improved endotoxin-induced ALI/ARDS and bleomycin-induced PF in mice. Thus, EnC- and EpC-II-derived EXOs regulate the immune balance of AMs and can be used as potential therapeutic drugs.
Collapse
Affiliation(s)
- Zunyong Feng
- grid.89957.3a0000 0000 9255 8984Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China ,grid.443626.10000 0004 1798 4069Department of Pathology, The First Affiliated Yijishan Hospital of Wannan Medical College & Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wannan Medical College, Wuhu, China ,grid.89957.3a0000 0000 9255 8984Interdisciplinary Innovation Institute for Medicine and Engineering, Southeast University-Nanjing Medical University, Nanjing, China
| | - Jing Zhou
- grid.443626.10000 0004 1798 4069Department of Pathology, The First Affiliated Yijishan Hospital of Wannan Medical College & Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wannan Medical College, Wuhu, China
| | - Yinhua Liu
- grid.443626.10000 0004 1798 4069Department of Pathology, The First Affiliated Yijishan Hospital of Wannan Medical College & Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wannan Medical College, Wuhu, China
| | - Ruixue Xia
- grid.459620.cDepartment of Respiratory and Critical Care Medicine, Henan University Huaihe Hospital, Kaifeng, China
| | - Qiang Li
- grid.443626.10000 0004 1798 4069Department of Anatomy, Wannan Medical College, Wuhu, China
| | - Liang Yan
- grid.443626.10000 0004 1798 4069Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Qun Chen
- grid.452929.1Department of Intensive Care Unit, Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Xiaobing Chen
- grid.414008.90000 0004 1799 4638Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuxin Jiang
- grid.411870.b0000 0001 0063 8301Department of Pathogenic Biology and Immunology, School of Medicine, Jiaxing University, Jiaxing, China
| | - Gao Chao
- grid.43169.390000 0001 0599 1243Department of Microsurgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ming Wang
- grid.216417.70000 0001 0379 7164Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guoren Zhou
- grid.452509.f0000 0004 1764 4566Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yijie Zhang
- grid.459620.cDepartment of Respiratory and Critical Care Medicine, Henan University Huaihe Hospital, Kaifeng, China
| | - Yongsheng Wang
- grid.428392.60000 0004 1800 1685Department of Respiratory Medicine, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hongping Xia
- grid.89957.3a0000 0000 9255 8984Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & State Key Laboratory of Reproductive Medicine & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China ,grid.443626.10000 0004 1798 4069Department of Pathology, The First Affiliated Yijishan Hospital of Wannan Medical College & Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, Wannan Medical College, Wuhu, China ,grid.89957.3a0000 0000 9255 8984Interdisciplinary Innovation Institute for Medicine and Engineering, Southeast University-Nanjing Medical University, Nanjing, China ,grid.459620.cDepartment of Respiratory and Critical Care Medicine, Henan University Huaihe Hospital, Kaifeng, China ,grid.452509.f0000 0004 1764 4566Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
24
|
Batista LFS, Torrecilha RBP, Silva RB, Utsunomiya YT, Silva TBF, Tomokane TY, Pacheco AD, Bosco AM, Paulan SC, Rossi CN, Costa GNO, Marcondes M, Ciarlini PC, Nunes CM, Matta VLR, Laurenti MD. Chromosomal segments may explain the antibody response cooperation for canine leishmaniasis pathogenesis. Vet Parasitol 2020; 288:109276. [PMID: 33152678 DOI: 10.1016/j.vetpar.2020.109276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/22/2022]
Abstract
Visceral leishmaniasis (VL) is marked by hyperactivation of a humoral response secreting high quantity of immunoglobulins (Igs) that are inaccessible to intracellular parasites. Here we investigated the contributions of the antibody response to the canine leishmaniasis pathogenesis. Using correlation and genome-wide association analysis, we investigated the relationship of anti-Leishmania infantum immunoglobulin classes levels with parasite burden, clinical response, renal/hepatic biochemical, and oxidative stress markers in dogs from endemic areas of VL. Immunoglobulin G (IgG) and IgA were positively correlated with parasite burden on lymph node and blood. Increased IgG, IgA and IgE levels were associated with severe canine leishmaniasis (CanL) whereas IgM was elevated in uninfected exposed dogs. Correlations of IgM, IgG and IgA with creatinine, urea, AST and ALT levels in the serum were suggested an involvement of those Igs with renal and hepatic changes. The correlogram of oxidative radicals and antioxidants revealed a likely relationship of IgM, IgG and IgA with oxidative stress and lipid peroxidation in the blood, suggested as mechanisms mediating tissue damage and CanL worsening. The gene mapping on chromosomal segments associated with the quantitative variation of immunoglobulin classes identified genetic signatures involved with reactive oxygen species generation, phagolysosome maturation and rupture, free iron availability, Th1/Th2 differenciation and, immunoglobulin clearance. The findings demonstrated the roles of the antibody response as resistance or susceptibility markers and mediators of CanL pathogenesis. In addition we pinpointed candidate genes as potential targets for the therapy against the damage caused by exacerbated antibody response and parasitism in VL.
Collapse
Affiliation(s)
- Luís F S Batista
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| | - Rafaela B P Torrecilha
- Departamento De Medicina Veterinária Preventiva e Reprodução Animal, Faculdade De Ciências Agrárias e Veterinárias, Univ Estadual Paulista, Jaboticabal, São Paulo, CEP: 14884-900, Brazil.
| | - Rafaela B Silva
- Escola de Saúde, Universidade Salvador, Salvador, Bahia, CEP: 41720-200, Brazil.
| | - Yuri T Utsunomiya
- Departamento de Apoio, Produção e Saúde Animal, Faculdade de Medicina Veterinária de Araçatuba, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Thaís B F Silva
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| | - Thaíse Y Tomokane
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| | - Acácio D Pacheco
- Departamento de Clínica, Cirurgia e Reprodução Animal, Faculdade de Medicina Veterinária, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Anelise M Bosco
- Departamento de Clínica, Cirurgia e Reprodução Animal, Faculdade de Medicina Veterinária, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Silvana C Paulan
- Departamento de Apoio, Produção e Saúde Animal, Faculdade de Medicina Veterinária de Araçatuba, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Claudio N Rossi
- Departamento de Clínica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, CEP 05508-270, Brazil.
| | - Gustavo N O Costa
- Departamento De Medicina Veterinária Preventiva e Reprodução Animal, Faculdade De Ciências Agrárias e Veterinárias, Univ Estadual Paulista, Jaboticabal, São Paulo, CEP: 14884-900, Brazil.
| | - Mary Marcondes
- Departamento de Clínica, Cirurgia e Reprodução Animal, Faculdade de Medicina Veterinária, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Paulo C Ciarlini
- Departamento de Clínica, Cirurgia e Reprodução Animal, Faculdade de Medicina Veterinária, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Cáris M Nunes
- Departamento de Apoio, Produção e Saúde Animal, Faculdade de Medicina Veterinária de Araçatuba, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Vânia L R Matta
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| | - Márcia D Laurenti
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| |
Collapse
|
25
|
Parisis D, Chivasso C, Perret J, Soyfoo MS, Delporte C. Current State of Knowledge on Primary Sjögren's Syndrome, an Autoimmune Exocrinopathy. J Clin Med 2020; 9:E2299. [PMID: 32698400 PMCID: PMC7408693 DOI: 10.3390/jcm9072299] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is a chronic systemic autoimmune rheumatic disease characterized by lymphoplasmacytic infiltration of the salivary and lacrimal glands, whereby sicca syndrome and/or systemic manifestations are the clinical hallmarks, associated with a particular autoantibody profile. pSS is the most frequent connective tissue disease after rheumatoid arthritis, affecting 0.3-3% of the population. Women are more prone to develop pSS than men, with a sex ratio of 9:1. Considered in the past as innocent collateral passive victims of autoimmunity, the epithelial cells of the salivary glands are now known to play an active role in the pathogenesis of the disease. The aetiology of the "autoimmune epithelitis" still remains unknown, but certainly involves genetic, environmental and hormonal factors. Later during the disease evolution, the subsequent chronic activation of B cells can lead to the development of systemic manifestations or non-Hodgkin's lymphoma. The aim of the present comprehensive review is to provide the current state of knowledge on pSS. The review addresses the clinical manifestations and complications of the disease, the diagnostic workup, the pathogenic mechanisms and the therapeutic approaches.
Collapse
Affiliation(s)
- Dorian Parisis
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium; (D.P.); (C.C.); (J.P.)
- Department of Rheumatology, Erasme Hospital, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Clara Chivasso
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium; (D.P.); (C.C.); (J.P.)
| | - Jason Perret
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium; (D.P.); (C.C.); (J.P.)
| | | | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 1070 Brussels, Belgium; (D.P.); (C.C.); (J.P.)
| |
Collapse
|
26
|
Gil-Varea E, Spataro N, Villar LM, Tejeda-Velarde A, Midaglia L, Matesanz F, Malhotra S, Eixarch H, Patsopoulos N, Fernández Ó, Oliver-Martos B, Saiz A, Llufriu S, Ramió-Torrentà L, Quintana E, Izquierdo G, Alcina A, Bosch E, Navarro A, Montalban X, Comabella M. Targeted resequencing reveals rare variants enrichment in multiple sclerosis susceptibility genes. Hum Mutat 2020; 41:1308-1320. [PMID: 32196808 DOI: 10.1002/humu.24016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 03/05/2020] [Accepted: 03/18/2020] [Indexed: 12/25/2022]
Abstract
Although genome-wide association studies have identified a number of common variants associated with multiple sclerosis (MS) susceptibility, little is known about the relevance of rare variants. Here, we aimed to explore the role of rare variants in 14 MS risk genes (FCRL1, RGS1, TIMMDC1, HHEX, CXCR5, LTBR, TSFM, GALC, TRAF3, STAT3, TNFSF14, IFI30, CD40, and CYP24A1) by targeted resequencing in an Iberian population of 524 MS cases and 546 healthy controls. Four rare variants-enriched regions within CYP24A1, FCRL1, RGS1, and TRAF3 were identified as significantly associated with MS. Functional studies revealed significantly decreased regulator of G protein signaling 1 (RGS1) gene expression levels in peripheral blood mononuclear cells from MS patients with RGS1 rare variants compared to noncarriers, whereas no significant differences in gene expression were observed for CYP24A1, FCRL1, and TRAF3 between rare variants carriers and noncarriers. Immunophenotyping showed significant decrease in RGS1 expression in peripheral blood B lymphocytes from MS patients with RGS1 rare variants relative to noncarriers. Lastly, peripheral blood mononuclear cell from MS patients carrying RGS1 rare variants showed significantly lower induction of RGS1 gene expression by interferon-β compared to MS patients lacking RGS1 variants. The presence of rare variants in RGS1 reinforce the ideas of high genetic heterogeneity and a role of rare variants in MS pathogenesis.
Collapse
Affiliation(s)
- Elia Gil-Varea
- Servei de Neurologia-Neuroimmunologia, Center d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nino Spataro
- Genetics Laboratory, UDIAT-Centre Diagnòstic, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Luisa María Villar
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | - Amalia Tejeda-Velarde
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | - Luciana Midaglia
- Servei de Neurologia-Neuroimmunologia, Center d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fuencisla Matesanz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Sunny Malhotra
- Servei de Neurologia-Neuroimmunologia, Center d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Center d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nikolaos Patsopoulos
- Department of Neurology, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Óscar Fernández
- Neuroimmunology and Neuroinflammation Group, Instituto de Investigación Biomédica de Málaga-IBIMA. UGC Neurociencias, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Begoña Oliver-Martos
- Neuroimmunology and Neuroinflammation Group, Instituto de Investigación Biomédica de Málaga-IBIMA. UGC Neurociencias, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Albert Saiz
- Servicio de Neurología, Hospital Clinic and Institut d'Investigació Biomèdica Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Llufriu
- Servicio de Neurología, Hospital Clinic and Institut d'Investigació Biomèdica Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lluís Ramió-Torrentà
- Department of Medical Sciences, Faculty of Medicine, Neurodegeneration and Neuroinflammation Group, Girona Biomedical Research Institute (IdIBGi), University of Girona, Girona, Spain
| | - Ester Quintana
- Department of Medical Sciences, Faculty of Medicine, Neurodegeneration and Neuroinflammation Group, Girona Biomedical Research Institute (IdIBGi), University of Girona, Girona, Spain
| | - Guillermo Izquierdo
- Departamento de Neurología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Antonio Alcina
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Elena Bosch
- Genetics Laboratory, UDIAT-Centre Diagnòstic, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Reus, Spain
| | - Arcadi Navarro
- Genetics Laboratory, UDIAT-Centre Diagnòstic, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain.,Centre de Regulació Genòmica (CRG), Barcelona, España.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Cataluña, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Center d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Center d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Lämmermann T, Kastenmüller W. Concepts of GPCR-controlled navigation in the immune system. Immunol Rev 2020; 289:205-231. [PMID: 30977203 PMCID: PMC6487968 DOI: 10.1111/imr.12752] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/01/2019] [Accepted: 02/03/2019] [Indexed: 12/11/2022]
Abstract
G‐protein–coupled receptor (GPCR) signaling is essential for the spatiotemporal control of leukocyte dynamics during immune responses. For efficient navigation through mammalian tissues, most leukocyte types express more than one GPCR on their surface and sense a wide range of chemokines and chemoattractants, leading to basic forms of leukocyte movement (chemokinesis, haptokinesis, chemotaxis, haptotaxis, and chemorepulsion). How leukocytes integrate multiple GPCR signals and make directional decisions in lymphoid and inflamed tissues is still subject of intense research. Many of our concepts on GPCR‐controlled leukocyte navigation in the presence of multiple GPCR signals derive from in vitro chemotaxis studies and lower vertebrates. In this review, we refer to these concepts and critically contemplate their relevance for the directional movement of several leukocyte subsets (neutrophils, T cells, and dendritic cells) in the complexity of mouse tissues. We discuss how leukocyte navigation can be regulated at the level of only a single GPCR (surface expression, competitive antagonism, oligomerization, homologous desensitization, and receptor internalization) or multiple GPCRs (synergy, hierarchical and non‐hierarchical competition, sequential signaling, heterologous desensitization, and agonist scavenging). In particular, we will highlight recent advances in understanding GPCR‐controlled leukocyte navigation by intravital microscopy of immune cells in mice.
Collapse
Affiliation(s)
- Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | |
Collapse
|
28
|
Mcheik S, Van Eeckhout N, De Poorter C, Galés C, Parmentier M, Springael JY. Coexpression of CCR7 and CXCR4 During B Cell Development Controls CXCR4 Responsiveness and Bone Marrow Homing. Front Immunol 2019; 10:2970. [PMID: 31921208 PMCID: PMC6930800 DOI: 10.3389/fimmu.2019.02970] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022] Open
Abstract
The CXCL12-CXCR4 axis plays a key role in the retention of stem cells and progenitors in dedicated bone marrow niches. It is well-known that CXCR4 responsiveness in B lymphocytes decreases dramatically during the final stages of their development in the bone marrow. However, the molecular mechanism underlying this regulation and whether it plays a role in B-cell homeostasis remain unknown. In the present study, we show that the differentiation of pre-B cells into immature and mature B cells is accompanied by modifications to the relative expression of chemokine receptors, with a two-fold downregulation of CXCR4 and upregulation of CCR7. We demonstrate that expression of CCR7 in B cells is involved in the selective inactivation of CXCR4, and that mature B cells from CCR7-/- mice display higher responsiveness to CXCL12 and improved retention in the bone marrow. We also provide molecular evidence supporting a model in which upregulation of CCR7 favors the formation of CXCR4-CCR7 heteromers, wherein CXCR4 is selectively impaired in its ability to activate certain G-protein complexes. Collectively, our results demonstrate that CCR7 behaves as a novel selective endogenous allosteric modulator of CXCR4.
Collapse
Affiliation(s)
- Saria Mcheik
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Nils Van Eeckhout
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Cédric De Poorter
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Céline Galés
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Marc Parmentier
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
| | - Jean-Yves Springael
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| |
Collapse
|
29
|
Dolcino M, Tinazzi E, Vitali C, Del Papa N, Puccetti A, Lunardi C. Long Non-Coding RNAs Modulate Sjögren's Syndrome Associated Gene Expression and Are Involved in the Pathogenesis of the Disease. J Clin Med 2019; 8:jcm8091349. [PMID: 31480511 PMCID: PMC6780488 DOI: 10.3390/jcm8091349] [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: 07/31/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023] Open
Abstract
Primary Sjögren's syndrome (pSjS) is a chronic systemic autoimmune disorder, primarily affecting exocrine glands; its pathogenesis is still unclear. Long non-coding RNAs (lncRNAs) are thought to play a role in the pathogenesis of autoimmune diseases and a comprehensive analysis of lncRNAs expression in pSjS is still lacking. To this aim, the expression of more than 540,000 human transcripts, including those ascribed to more than 50,000 lncRNAs is profiled at the same time, in a cohort of 16 peripheral blood mononuclear cells PBMCs samples (eight pSjS and eight healthy subjects). A complex network analysis is carried out on the global set of molecular interactions among modulated genes and lncRNAs, leading to the identification of reliable lncRNA-miRNA-gene functional interactions. Taking this approach, a few lncRNAs are identified as targeting highly connected genes in the pSjS transcriptome, since they have a major impact on gene modulation in the disease. Such genes are involved in biological processes and molecular pathways crucial in the pathogenesis of pSjS, including immune response, B cell development and function, inflammation, apoptosis, type I and gamma interferon, epithelial cell adhesion and polarization. The identification of deregulated lncRNAs that modulate genes involved in the typical features of the disease provides insight in disease pathogenesis and opens avenues for the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Marzia Dolcino
- Department of Medicine, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Elisa Tinazzi
- Department of Medicine, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Claudio Vitali
- Sections of Rheumatology, Villa S. Giuseppe, Como and Casa di Cura di Lecco, 23900 Lecco, Italy
| | | | - Antonio Puccetti
- Department of Experimental Medicine, Section of Histology, University of Genova, Via G.B. Marsano 10, 16132 Genova, Italy
| | - Claudio Lunardi
- Department of Medicine, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy.
| |
Collapse
|
30
|
Zhu Y, Zhao Y, Dong S, Liu L, Tai L, Xu Y. Systematic identification of dysregulated lncRNAs associated with platinum-based chemotherapy response across 11 cancer types. Genomics 2019; 112:1214-1222. [PMID: 31302201 DOI: 10.1016/j.ygeno.2019.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/26/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
Aberrant expression of long non-coding RNAs (lncRNAs) leads to the development of chemoresistance by regulating a series of biological processes, which is one of the major obstacles in the cancer treatment. This study aimed to identify some key lncRNAs that are associated with platinum-based chemoresistance in multiple cancers. Regulating the expression levels of these lncRNAs can enhance the sensitivity of patients to chemotherapy drugs and improve the therapeutic effect of cancer. By systematically analyzing 648 samples regarding platinum drug response from the Cancer Genome Atlas (TCGA), we have identified 32 dysregulated lncRNAs across 11 cancer types that could affect platinum-based chemotherapy response, of which 78.125% (25/32) were significantly down-regulated in drug-resistant samples. Drug response prediction model that had been constructed based on the expression pattern of these dysregulated lncRNAs could accurately predict the chemotherapy response of tumor patients, and the area under the curve (AUC) was between 0.8034 and 0.9984. In particular, all of these dysregulated lncRNAs that we identified were cancer-specific. They were significantly associated with the survival of tumor patients and could serve as cancer-specific biomarkers for prognosis. In conclusion, this study will contribute to improving the drug resistance of tumor patients during chemotherapy, and it is of real significance for selecting effective chemotherapy drugs and achieving precision medicine.
Collapse
Affiliation(s)
- Yanjiao Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yichuan Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Siyao Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lu Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lin Tai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| |
Collapse
|
31
|
Sun Q, He Q, Xu J, Liu Q, Lu Y, Zhang Z, Xu X, Sun B. Guanine nucleotide-binding protein G(i)α2 aggravates hepatic ischemia-reperfusion injury in mice by regulating MLK3 signaling. FASEB J 2019; 33:7049-7060. [PMID: 30840837 DOI: 10.1096/fj.201802462r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Hepatic ischemia-reperfusion (I/R) injury is a major challenge in liver resection and transplantation surgeries. Previous studies have revealed that guanine nucleotide-binding protein G(i)α2 (GNAI2) was involved in the progression of myocardial and cerebral I/R injury, but the role and function of GNAI2 in hepatic I/R have not been elucidated. The hepatocyte-specific GNAI2 knockout (GNAI2hep-/-) mice were generated and subjected to hepatic I/R injury. Primary hepatocytes isolated from GNAI2hep-/- and GNAI2flox/flox mice were cultured and challenged to hypoxia-reoxygenation insult. The specific function of GNAI2 in I/R-triggered hepatic injury and the underlying molecular mechanism were explored by various phenotypic analyses and molecular biology methods. In this study, we demonstrated that hepatic GNAI2 expression was significantly increased in liver transplantation patients and wild-type mice after hepatic I/R. Interestingly, hepatocyte-specific GNAI2 deficiency attenuated I/R-induced liver damage, inflammation cytokine expression, macrophage/neutrophil infiltration, and hepatocyte apoptosis in vivo and in vitro. Mechanistically, up-regulation of GNAI2 phosphorylates mixed-lineage protein kinase 3 (MLK3) through direct binding, which exacerbated hepatic I/R damage via MAPK and NF-κB pathway activation. Furthermore, blocking MLK3 signaling reversed GNAI2-mediated hepatic I/R injury. Our study firstly identifies GNAI2 as a promising target for prevention of hepatic I/R-induced injury and related liver diseases.-Sun, Q., He, Q., Xu, J., Liu, Q., Lu, Y., Zhang, Z., Xu, X., Sun, B. Guanine nucleotide-binding protein G(i)α2 aggravates hepatic ischemia-reperfusion injury in mice by regulating MLK3 signaling.
Collapse
Affiliation(s)
- Qikai Sun
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; and
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qifeng He
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; and
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jianbo Xu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; and
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiaoyu Liu
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; and
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yijun Lu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; and
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zechuan Zhang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; and
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaoliang Xu
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; and
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Beicheng Sun
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; and
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
32
|
Brink T, Leiss V, Siegert P, Jehle D, Ebner JK, Schwan C, Shymanets A, Wiese S, Nürnberg B, Hensel M, Aktories K, Orth JHC. Salmonella Typhimurium effector SseI inhibits chemotaxis and increases host cell survival by deamidation of heterotrimeric Gi proteins. PLoS Pathog 2018; 14:e1007248. [PMID: 30102745 PMCID: PMC6107295 DOI: 10.1371/journal.ppat.1007248] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/23/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022] Open
Abstract
Salmonella enterica serotype Typhimurium (S. Typhimurium) is one of the most frequent causes of food-borne illness in humans and usually associated with acute self-limiting gastroenteritis. However, in immunocompromised patients, the pathogen can disseminate and lead to severe systemic diseases. S. Typhimurium are facultative intracellular bacteria. For uptake and intracellular life, Salmonella translocate numerous effector proteins into host cells using two type-III secretion systems (T3SS), which are encoded within Salmonella pathogenicity islands 1 (SPI-1) and 2 (SPI-2). While SPI-1 effectors mainly promote initial invasion, SPI-2 effectors control intracellular survival and proliferation. Here, we elucidate the mode of action of Salmonella SPI-2 effector SseI, which is involved in control of systemic dissemination of S. Typhimurium. SseI deamidates a specific glutamine residue of heterotrimeric G proteins of the Gαi family, resulting in persistent activation of the G protein. Gi activation inhibits cAMP production and stimulates PI3-kinase γ by Gαi-released Gβγ subunits, resulting in activation of survival pathways by phosphorylation of Akt and mTOR. Moreover, SseI-induced deamidation leads to non-polarized activation of Gαi and, thereby, to loss of directed migration of dendritic cells. Salmonella Typhimurium is one of the most common causes of gastroenteritis in humans. In immunocompromised patients, the pathogen can cause systemic infections. Crucial virulence factors are encoded on two Salmonella pathogenicity islands SPI-1 and SPI-2. While SPI-1 encodes virulence factors essential for host cell invasion, intracellular proliferation of the pathogen depends mainly on SPI-2 effectors. Here, we elucidate the mode of action of Salmonella SPI-2 effector SseI. SseI activates heterotrimeric G proteins of the Gαi family by deamidation of a specific glutamine residue. Deamidation blocks GTP hydrolysis by Gαi, resulting in a persistently active G protein. Gi activation inhibits cAMP production and stimulates PI3Kγ by Gαi-released Gβγ subunits, resulting in activation of survival pathways by phosphorylation of Akt and mTOR. Moreover, deamidation of Gαi leads to a loss of directed migration in dendritic cells. The data offers a new perspective in the understanding of the actions of SseI.
Collapse
Affiliation(s)
- Thorsten Brink
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Veronika Leiss
- Abteilung für Pharmakologie und Experimentelle Therapie, Medizinische Fakultät und ICePhA, Eberhard-Karls-Universität Tübingen, Germany
| | - Peter Siegert
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Doris Jehle
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Julia K. Ebner
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- Fakultät für Biologie, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Carsten Schwan
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Aliaksei Shymanets
- Abteilung für Pharmakologie und Experimentelle Therapie, Medizinische Fakultät und ICePhA, Eberhard-Karls-Universität Tübingen, Germany
| | - Sebastian Wiese
- Zentrum für Biosystemanalyse, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Bernd Nürnberg
- Abteilung für Pharmakologie und Experimentelle Therapie, Medizinische Fakultät und ICePhA, Eberhard-Karls-Universität Tübingen, Germany
| | - Michael Hensel
- Abteilung Mikrobiologie, Fachbereich Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany
| | - Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- * E-mail:
| | - Joachim H. C. Orth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Perschbacher KJ, Deng G, Fisher RA, Gibson-Corley KN, Santillan MK, Grobe JL. Regulators of G protein signaling in cardiovascular function during pregnancy. Physiol Genomics 2018; 50:590-604. [PMID: 29702036 PMCID: PMC6139632 DOI: 10.1152/physiolgenomics.00037.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptor signaling mechanisms are implicated in many aspects of cardiovascular control, and dysfunction of such signaling mechanisms is commonly associated with disease states. Investigators have identified a large number of regulator of G protein signaling (RGS) proteins that variously contribute to the modulation of intracellular second-messenger signaling kinetics. These many RGS proteins each interact with a specific set of second-messenger cascades and receptor types and exhibit tissue-specific expression patterns. Increasing evidence supports the contribution of RGS proteins, or their loss, in the pathogenesis of cardiovascular dysfunctions. This review summarizes the current understanding of the functional contributions of RGS proteins, particularly within the B/R4 family, in cardiovascular disorders of pregnancy including gestational hypertension, uterine artery dysfunction, and preeclampsia.
Collapse
Affiliation(s)
| | - Guorui Deng
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | - Rory A Fisher
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | - Katherine N Gibson-Corley
- Department of Pathology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
| | - Mark K Santillan
- Department of Obstetrics & Gynecology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
- Fraternal Order of Eagles' Diabetes Research Center, University of Iowa , Iowa City, Iowa
- Obesity Education & Research Initiative, University of Iowa , Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa , Iowa City, Iowa
| |
Collapse
|
34
|
Laufer JM, Legler DF. Beyond migration-Chemokines in lymphocyte priming, differentiation, and modulating effector functions. J Leukoc Biol 2018; 104:301-312. [PMID: 29668063 DOI: 10.1002/jlb.2mr1217-494r] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/08/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023] Open
Abstract
Chemokines and their receptors coordinate the positioning of leukocytes, and lymphocytes in particular, in space and time. Discrete lymphocyte subsets, depending on their activation and differentiation status, express various sets of chemokine receptors to be recruited to distinct tissues. Thus, the network of chemokines and their receptors ensures the correct localization of specialized lymphocyte subsets within the appropriate microenvironment enabling them to search for cognate antigens, to become activated, and to fulfill their effector functions. The chemokine system therefore is vital for the initiation as well as the regulation of immune responses to protect the body from pathogens while maintaining tolerance towards self. Besides the well investigated function of orchestrating directed cell migration, chemokines additionally act on lymphocytes in multiple ways to shape immune responses. In this review, we highlight and discuss the role of chemokines and chemokine receptors in controlling cell-to-cell contacts required for lymphocyte arrest on endothelial cells and immunological synapse formation, in lymphocyte priming and differentiation, survival, as well as in modulating effector functions.
Collapse
Affiliation(s)
- Julia M Laufer
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland.,Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland.,Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
35
|
Park C, Hwang IY, Kehrl JH. The Use of Intravital Two-Photon and Thick Section Confocal Imaging to Analyze B Lymphocyte Trafficking in Lymph Nodes and Spleen. Methods Mol Biol 2018; 1707:193-205. [PMID: 29388109 DOI: 10.1007/978-1-4939-7474-0_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Intravital two-photon laser scanning microscopy (TP-LSM) has allowed the direct observation of immune cells in intact organs of living animals. In the B cell biology field TP-LSM has detailed the movement of B cells in high endothelial venules and during their transmigration into lymph organs; described the movement and positioning of B cells within lymphoid organs; outlined the mechanisms by which antigen is delivered to B cells; observed B cell interacting with T cells, other cell types, and even with pathogens; and delineated the egress of B cells from the lymph node (LN) parenchyma into the efferent lymphatics. As the quality of TP-LSM improves and as new fluorescent probes become available additional insights into B cell behavior and function await new investigations. Yet intravital TP-LSM has some disadvantages including a lower resolution than standard confocal microscopy, a narrow imaging window, and a shallow depth of imaging. We have found that supplementing intravital TP-LSM with conventional confocal microscopy using thick LN sections helps to overcome some of these shortcomings. Here, we describe procedures for visualizing the behavior and trafficking of fluorescently labeled, adoptively transferred antigen-activated B cells within the inguinal LN of live mice using two-photon microscopy. Also, we introduce procedures for fixed thick section imaging using standard confocal microscopy, which allows imaging of fluorescently labeled cells deep in the LN cortex and in the spleen with high resolution.
Collapse
Affiliation(s)
- Chung Park
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Il-Young Hwang
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - John H Kehrl
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
36
|
Park JH, Kim JH, Jo KE, Na SW, Eisenhut M, Kronbichler A, Lee KH, Shin JI. Field Synopsis and Re-analysis of Systematic Meta-analyses of Genetic Association Studies in Multiple Sclerosis: a Bayesian Approach. Mol Neurobiol 2017; 55:5672-5688. [PMID: 29027112 DOI: 10.1007/s12035-017-0773-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 09/12/2017] [Indexed: 12/31/2022]
Abstract
To provide an up-to-date summary of multiple sclerosis-susceptible gene variants and assess the noteworthiness in hopes of finding true associations, we investigated the results of 44 meta-analyses on gene variants and multiple sclerosis published through December 2016. Out of 70 statistically significant genotype associations, roughly a fifth (21%) of the comparisons showed noteworthy false-positive rate probability (FPRP) at a statistical power to detect an OR of 1.5 and at a prior probability of 10-6 assumed for a random single nucleotide polymorphism. These associations (IRF8/rs17445836, STAT3/rs744166, HLA/rs4959093, HLA/rs2647046, HLA/rs7382297, HLA/rs17421624, HLA/rs2517646, HLA/rs9261491, HLA/rs2857439, HLA/rs16896944, HLA/rs3132671, HLA/rs2857435, HLA/rs9261471, HLA/rs2523393, HLA-DRB1/rs3135388, RGS1/rs2760524, PTGER4/rs9292777) also showed a noteworthy Bayesian false discovery probability (BFDP) and one additional association (CD24 rs8734/rs52812045) was also noteworthy via BFDP computation. Herein, we have identified several noteworthy biomarkers of multiple sclerosis susceptibility. We hope these data are used to study multiple sclerosis genetics and inform future screening programs.
Collapse
Affiliation(s)
- Jae Hyon Park
- Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joo Hi Kim
- Yonsei University Wonju College of Medicine, Seoul, Republic of Korea
| | - Kye Eun Jo
- College of Medicine, University of Debrecen, Debrecen, Hungary
| | - Se Whan Na
- Yonsei University Wonju College of Medicine, Seoul, Republic of Korea
| | - Michael Eisenhut
- Department of Pediatrics, Luton & Dunstable University Hospital NHS Foundation Trust, Luton, UK
| | - Andreas Kronbichler
- Department of Internal Medicine IV, Medical University Innsbruck, Innsbruck, Austria
| | - Keum Hwa Lee
- Department of Pediatrics, Yonsei University College of Medicine, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, Republic of Korea
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, Republic of Korea.
| |
Collapse
|
37
|
Lee HK, Kim HS, Kim JS, Kim YG, Park KH, Lee JH, Kim KH, Chang IY, Bae SC, Kim Y, Hong JT, Kehrl JH, Han SB. CCL2 deficient mesenchymal stem cells fail to establish long-lasting contact with T cells and no longer ameliorate lupus symptoms. Sci Rep 2017; 7:41258. [PMID: 28117437 PMCID: PMC5259742 DOI: 10.1038/srep41258] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease characterized by autoantibody production. Mesenchymal stem cells (MSCs) ameliorate SLE symptoms by targeting T cells, whereas the mechanisms of their efficacy remain incompletely understood. In this study, we show that transfer of human MSCs increased MRL.Faslpr mouse survival, decreased T cell infiltration in the kidneys, and reduced T cell cytokine expression. In vitro, allogeneic mouse MSCs inhibited MRL.Faslpr T cell proliferation and cytokine production. Time-lapse imaging revealed that MSCs recruited MRL.Faslpr T cells establishing long-lasting cellular contacts by enhancing T cell VCAM-1 expression in a CCL2-dependent manner. In contrast, CCL2 deficient MSCs did not induce T cell migration and VCAM-1 expression, resulting in insufficient cell-cell contact. Consequently, CCL2 deficient MSCs did not inhibit IFN-γ production by T cells and upon transfer no longer prolonged survival of MRL.Faslpr mice. Taken together, our imaging study demonstrates that CCL2 enables the prolonged MSC-T cell interactions needed for sufficient suppression of autoreactive T cells and helps to understand how MSCs ameliorate symptoms in lupus-prone MRL.Faslpr mice.
Collapse
Affiliation(s)
- Hong Kyung Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Hyung Sook Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea.,Corestem Inc, Gyeonggi 13486, Republic of Korea
| | - Ji Sung Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Yong Guk Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Ki Hwan Park
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jae Hee Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Ki Hun Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | | | - Sang-Cheol Bae
- Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Republic of Korea
| | - Youngsoo Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - John H Kehrl
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| |
Collapse
|
38
|
Robichaux WG, Branham-O'Connor M, Hwang IY, Vural A, Kehrl JH, Blumer JB. Regulation of Chemokine Signal Integration by Activator of G-Protein Signaling 4 (AGS4). J Pharmacol Exp Ther 2017; 360:424-433. [PMID: 28062526 DOI: 10.1124/jpet.116.238436] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 12/28/2016] [Indexed: 12/15/2022] Open
Abstract
Activator of G-protein signaling 4 (AGS4)/G-protein signaling modulator 3 (Gpsm3) contains three G-protein regulatory (GPR) motifs, each of which can bind Gαi-GDP free of Gβγ We previously demonstrated that the AGS4-Gαi interaction is regulated by seven transmembrane-spanning receptors (7-TMR), which may reflect direct coupling of the GPR-Gαi module to the receptor analogous to canonical Gαβγ heterotrimer. We have demonstrated that the AGS4-Gαi complex is regulated by chemokine receptors in an agonist-dependent manner that is receptor-proximal. As an initial approach to investigate the functional role(s) of this regulated interaction in vivo, we analyzed leukocytes, in which AGS4/Gpsm3 is predominantly expressed, from AGS4/Gpsm3-null mice. Loss of AGS4/Gpsm3 resulted in mild but significant neutropenia and leukocytosis. Dendritic cells, T lymphocytes, and neutrophils from AGS4/Gpsm3-null mice also exhibited significant defects in chemoattractant-directed chemotaxis and extracellular signal-regulated kinase activation. An in vivo peritonitis model revealed a dramatic reduction in the ability of AGS4/Gpsm3-null neutrophils to migrate to primary sites of inflammation. Taken together, these data suggest that AGS4/Gpsm3 is required for proper chemokine signal processing in leukocytes and provide further evidence for the importance of the GPR-Gαi module in the regulation of leukocyte function.
Collapse
Affiliation(s)
- William G Robichaux
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina (W.G.R., M.B.-O., J.B.B.); and B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (I.-Y.H., A.V., J.H.K.)
| | - Melissa Branham-O'Connor
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina (W.G.R., M.B.-O., J.B.B.); and B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (I.-Y.H., A.V., J.H.K.)
| | - Il-Young Hwang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina (W.G.R., M.B.-O., J.B.B.); and B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (I.-Y.H., A.V., J.H.K.)
| | - Ali Vural
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina (W.G.R., M.B.-O., J.B.B.); and B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (I.-Y.H., A.V., J.H.K.)
| | - Johne H Kehrl
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina (W.G.R., M.B.-O., J.B.B.); and B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (I.-Y.H., A.V., J.H.K.)
| | - Joe B Blumer
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina (W.G.R., M.B.-O., J.B.B.); and B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (I.-Y.H., A.V., J.H.K.)
| |
Collapse
|
39
|
Druey KM. Emerging Roles of Regulators of G Protein Signaling (RGS) Proteins in the Immune System. Adv Immunol 2017; 136:315-351. [PMID: 28950950 DOI: 10.1016/bs.ai.2017.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Kirk M Druey
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States.
| |
Collapse
|
40
|
Arvind P, Jayashree S, Jambunathan S, Nair J, Kakkar VV. Understanding gene expression in coronary artery disease through global profiling, network analysis and independent validation of key candidate genes. J Genet 2016; 94:601-10. [PMID: 26690514 DOI: 10.1007/s12041-015-0548-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Molecular mechanism underlying the patho-physiology of coronary artery disease (CAD) is complex. We used global expression profiling combined with analysis of biological network to dissect out potential genes and pathways associated with CAD in a representative case-control Asian Indian cohort. We initially performed blood transcriptomics profiling in 20 subjects, including 10 CAD patients and 10 healthy controls on the Agilent microarray platform. Data was analysed with Gene Spring Gx12.5, followed by network analysis using David v 6.7 and Reactome databases. The most significant differentially expressed genes from microarray were independently validated by real time PCR in 97 cases and 97 controls. A total of 190 gene transcripts showed significant differential expression (fold change>2,P<0.05) between the cases and the controls of which 142 genes were upregulated and 48 genes were downregulated. Genes associated with inflammation, immune response, cell regulation, proliferation and apoptotic pathways were enriched, while inflammatory and immune response genes were displayed as hubs in the network, having greater number of interactions with the neighbouring genes. Expression of EGR1/2/3, IL8, CXCL1, PTGS2, CD69, IFNG, FASLG, CCL4, CDC42, DDX58, NFKBID and NR4A2 genes were independently validated; EGR1/2/3 and IL8 showed >8-fold higher expression in cases relative to the controls implying their important role in CAD. In conclusion, global gene expression profiling combined with network analysis can help in identifying key genes and pathways for CAD.
Collapse
Affiliation(s)
- Prathima Arvind
- Mary and Garry Weston Functional Genomics Unit, Thrombosis Research Institute, Bengaluru 560 099, India.
| | | | | | | | | |
Collapse
|
41
|
Doedens AL, Rubinstein MP, Gross ET, Best JA, Craig DH, Baker MK, Cole DJ, Bui JD, Goldrath AW. Molecular Programming of Tumor-Infiltrating CD8+ T Cells and IL15 Resistance. Cancer Immunol Res 2016; 4:799-811. [PMID: 27485135 PMCID: PMC5010943 DOI: 10.1158/2326-6066.cir-15-0178] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 06/29/2016] [Indexed: 12/24/2022]
Abstract
Despite clinical potential and recent advances, durable immunotherapeutic ablation of solid tumors is not routinely achieved. IL15 expands natural killer cell (NK), natural killer T cell (NKT) and CD8(+) T-cell numbers and engages the cytotoxic program, and thus is under evaluation for potentiation of cancer immunotherapy. We found that short-term therapy with IL15 bound to soluble IL15 receptor α-Fc (IL15cx; a form of IL15 with increased half-life and activity) was ineffective in the treatment of autochthonous PyMT murine mammary tumors, despite abundant CD8(+) T-cell infiltration. Probing of this poor responsiveness revealed that IL15cx only weakly activated intratumoral CD8(+) T cells, even though cells in the lung and spleen were activated and dramatically expanded. Tumor-infiltrating CD8(+) T cells exhibited cell-extrinsic and cell-intrinsic resistance to IL15. Our data showed that in the case of persistent viral or tumor antigen, single-agent systemic IL15cx treatment primarily expanded antigen-irrelevant or extratumoral CD8(+) T cells. We identified exhaustion, tissue-resident memory, and tumor-specific molecules expressed in tumor-infiltrating CD8(+) T cells, which may allow therapeutic targeting or programming of specific subsets to evade loss of function and cytokine resistance, and, in turn, increase the efficacy of IL2/15 adjuvant cytokine therapy. Cancer Immunol Res; 4(9); 799-811. ©2016 AACR.
Collapse
Affiliation(s)
- Andrew L Doedens
- Division of Biological Sciences, University of California, San Diego, La Jolla, California
| | - Mark P Rubinstein
- Division of Biological Sciences, University of California, San Diego, La Jolla, California. Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Emilie T Gross
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - J Adam Best
- Division of Biological Sciences, University of California, San Diego, La Jolla, California
| | - David H Craig
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Megan K Baker
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - David J Cole
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Jack D Bui
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Ananda W Goldrath
- Division of Biological Sciences, University of California, San Diego, La Jolla, California.
| |
Collapse
|
42
|
Lee HK, Kim YG, Kim JS, Park EJ, Kim B, Park KH, Kang JS, Hong JT, Kim Y, Han SB. Cytokine-induced killer cells interact with tumor lysate-pulsed dendritic cells via CCR5 signaling. Cancer Lett 2016; 378:142-9. [PMID: 27216980 DOI: 10.1016/j.canlet.2016.05.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 01/22/2023]
Abstract
The antitumor activity of cytokine-induced killer (CIK) cells can be increased by co-culturing them with tumor lysate-pulsed dendritic cells (tDCs); this phenomenon has been studied mainly at the population level. Using time-lapse imaging, we examined how CIK cells gather information from tDCs at the single-cell level. tDCs highly expressed CCL5, which bound CCR5 expressed on CIK cells. tDCs strongly induced migration of Ccr5(+/+) CIK cells, but not that of Ccr5(-/-) CIK cells or Ccr5(+/+) CIK cells treated with the CCR5 antagonist Maraviroc. Individual tDCs contacted Ccr5(+/+) CIK cells more frequently and lengthily than with Ccr5(-/-) CIK cells. Consequently, tDCs increased the antitumor activity of Ccr5(+/+) CIK cells in vitro and in vivo, but did not increase that of Ccr5(-/-) CIK cells. Taken together, our data provide insight into the mechanism of CIK cell activation by tDCs at the single-cell level.
Collapse
Affiliation(s)
- Hong Kyung Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Yong Guk Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Ji Sung Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Eun Jae Park
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Boyeong Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Ki Hwan Park
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Jong Soon Kang
- Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungbuk 28116, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Youngsoo Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea.
| |
Collapse
|
43
|
Kehrl JH. The impact of RGS and other G-protein regulatory proteins on Gαi-mediated signaling in immunity. Biochem Pharmacol 2016; 114:40-52. [PMID: 27071343 DOI: 10.1016/j.bcp.2016.04.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/08/2016] [Indexed: 01/30/2023]
Abstract
Leukocyte chemoattractant receptors are members of the G-protein coupled receptor (GPCR) family. Signaling downstream of these receptors directs the localization, positioning and homeostatic trafficking of leukocytes; as well as their recruitment to, and their retention at, inflammatory sites. Ligand induced changes in the molecular conformation of chemoattractant receptors results in the engagement of heterotrimeric G-proteins, which promotes α subunits to undergo GTP/GDP exchange. This results in the functional release of βγ subunits from the heterotrimers, thereby activating downstream effector molecules, which initiate leukocyte polarization, gradient sensing, and directional migration. Pertussis toxin ADP ribosylates Gαi subunits and prevents chemoattractant receptors from triggering Gαi nucleotide exchange. The use of pertussis toxin revealed the essential importance of Gαi subunit nucleotide exchange for chemoattractant receptor signaling. More recent studies have identified a range of regulatory mechanisms that target these receptors and their associated heterotrimeric G-proteins, thereby helping to control the magnitude, kinetics, and duration of signaling. A failure in these regulatory pathways can lead to impaired receptor signaling and immunopathology. The analysis of mice with targeted deletions of Gαi isoforms as well as some of these G-protein regulatory proteins is providing insights into their roles in chemoattractant receptor signaling.
Collapse
Affiliation(s)
- John H Kehrl
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 2089, United States.
| |
Collapse
|
44
|
The autoimmunity-associated gene RGS1 affects the frequency of T follicular helper cells. Genes Immun 2016; 17:228-38. [PMID: 27029527 PMCID: PMC4892947 DOI: 10.1038/gene.2016.16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/19/2016] [Accepted: 02/25/2016] [Indexed: 12/14/2022]
Abstract
RGS1 (regulator of G-protein signaling 1) has been associated with multiple autoimmune disorders including type I diabetes. RGS1 desensitizes the chemokine receptors CCR7 and CXCR4 that are critical to the localization of T and B cells in lymphoid organs. To explore how RGS1 variation contributes to autoimmunity, we generated Rgs1 knockdown (KD) mice in the nonobese diabetic (NOD) model for type I diabetes. We found that Rgs1 KD increased the size of germinal centers, but decreased the frequency of T follicular helper (TFH) cells. We show that loss of Rgs1 in T cells had both a T cell-intrinsic effect on migration and TFH cell frequency, and an indirect effect on B-cell migration and germinal center formation. Notably, several recent publications described an increase in circulating TFH cells in patients with type I diabetes, suggesting this cell population is involved in pathogenesis. Though Rgs1 KD was insufficient to alter diabetes frequency in the NOD model, our findings raise the possibility that RGS1 plays a role in autoimmunity owing to its function in TFH cells. This mechanistic link, although speculative at this time, would lend support to the notion that TFH cells are key participants in autoimmunity and could explain the association of RGS1 with several immune-mediated diseases.
Collapse
|
45
|
Meta-Analysis on Associations of RGS1 and IL12A Polymorphisms with Celiac Disease Risk. Int J Mol Sci 2016; 17:457. [PMID: 27043536 PMCID: PMC4848913 DOI: 10.3390/ijms17040457] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/17/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of celiac disease (CD) has been related to polymorphisms in the regulator of G-protein signaling 1 (RGS1) and interleukin-12 A (IL12A) genes, but the existing findings are inconsistent. Our aim is to investigate the associations of two single-nucleotide polymorphisms (SNPs) (rs2816316 in RGS1 and rs17810546 in IL12A) with CD risk using meta-analysis. We searched PubMed and Web of Science on RGS1 rs2816316 and IL12A rs17810546 with CD risk. Odds ratio (OR) and 95% confidence interval (CI) of each SNP were estimated. All statistical analyses were performed on Stata 12.0. A total of seven studies were retrieved and analyzed. The available data indicated the minor allele C of rs2816316 was negatively associated with CD (C vs. A: OR = 0.77, 95% CI = 0.74-0.80), and a positive association was found for the minor allele G of rs17810546 (G vs. A: OR = 1.37, 95% CI = 1.31-1.43). The co-dominant model of genotype effect confirmed the significant associations between RGS1 rs2816316/IL12A rs17810546 and CD. No evidence of publication bias was observed. Our meta-analysis supports the associations of RGS1 and IL12A with CD and strongly calls for further studies to better understand the roles of RGS1 and IL12A in the pathogenesis of CD.
Collapse
|
46
|
Kuwano Y, Adler M, Zhang H, Groisman A, Ley K. Gαi2 and Gαi3 Differentially Regulate Arrest from Flow and Chemotaxis in Mouse Neutrophils. THE JOURNAL OF IMMUNOLOGY 2016; 196:3828-33. [PMID: 26976957 DOI: 10.4049/jimmunol.1500532] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 02/22/2016] [Indexed: 01/13/2023]
Abstract
Leukocyte recruitment to inflammation sites progresses in a multistep cascade. Chemokines regulate multiple steps of the cascade, including arrest, transmigration, and chemotaxis. The most important chemokine receptor in mouse neutrophils is CXCR2, which couples through Gαi2- and Gαi3-containing heterotrimeric G proteins. Neutrophils arrest in response to CXCR2 stimulation. This is defective in Gαi2-deficient neutrophils. In this study, we show that Gαi3-deficient neutrophils showed reduced transmigration but normal arrest in mice. We also tested Gαi2- or Gαi3-deficient neutrophils in a CXCL1 gradient generated by a microfluidic device. Gαi3-, but not Gαi2-, deficient neutrophils showed significantly reduced migration and directionality. This was confirmed in a model of sterile inflammation in vivo. Gαi2-, but not Gαi3-, deficient neutrophils showed decreased Ca(2+) flux in response to CXCR2 stimulation. Conversely, Gαi3-, but not Gαi2-, deficient neutrophils exhibited reduced AKT phosphorylation upon CXCR2 stimulation. We conclude that Gαi2 controls arrest and Gαi3 controls transmigration and chemotaxis in response to chemokine stimulation of neutrophils.
Collapse
Affiliation(s)
- Yoshihiro Kuwano
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Micha Adler
- Department of Physics, University of California, San Diego, La Jolla, CA 92093; and
| | - Hong Zhang
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Alex Groisman
- Department of Physics, University of California, San Diego, La Jolla, CA 92093; and
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
47
|
Park C, Hwang IY, Kehrl JH. Intravital Two-Photon Imaging of Lymphocytes Crossing High Endothelial Venules and Cortical Lymphatics in the Inguinal Lymph Node. Methods Mol Biol 2016; 1407:195-206. [PMID: 27271904 DOI: 10.1007/978-1-4939-3480-5_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Lymphocyte recirculation through lymph nodes (LNs) requires their crossing of endothelial barriers present in blood vessels and lymphatics by means of chemoattractant-triggered cell migration. The chemoattractant-chemoattractant receptor axes that predominately govern the trafficking of lymphocytes into, and out of, LNs are CCL19/CCR7 and sphingosine 1-phosphate (S1P)/S1P receptor 1 (S1PR1), respectively. Blood-borne lymphocytes downregulate S1PR1 and use CCR7 signaling to adhere to high endothelial venules (HEVs) for transmigration. During their LN residency, recirculating lymphocytes reacquire S1PR1 and attenuate their sensitivity to chemokines. Eventually lymphocytes exit the LN by entering the cortical or medullary lymphatics, a process that depends upon S1PR1 signaling. Upon entering into the lymph, lymphocytes lose their polarity, downregulate their sensitivity to S1P due to the high concentration of S1P, and upregulate their sensitivity to chemokines. However, many of the details of lymphocyte transmigration across endothelial barriers remain poorly understood. Intravital two-photon imaging with advanced microscope technologies not only allows the real-time observation of immune cells in intact LN of a live mouse, but also provides a means to monitor the interactions between circulating lymphocytes and stromal barriers. Here, we describe procedures to visualize lymphocytes engaging and crossing HEVs, and approaching and crossing the cortical lymphatic endothelium to enter the efferent lymph in live mice.
Collapse
Affiliation(s)
- Chung Park
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bldg 10, Room 11B08, 10 Center Dr. MSC 1876, Bethesda, MD, 20892, USA
| | - Il-Young Hwang
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bldg 10, Room 11B08, 10 Center Dr. MSC 1876, Bethesda, MD, 20892, USA
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bldg 10, Room 11B08, 10 Center Dr. MSC 1876, Bethesda, MD, 20892, USA.
| |
Collapse
|
48
|
Xie Z, Chan EC, Druey KM. R4 Regulator of G Protein Signaling (RGS) Proteins in Inflammation and Immunity. AAPS JOURNAL 2015; 18:294-304. [PMID: 26597290 DOI: 10.1208/s12248-015-9847-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/11/2015] [Indexed: 11/30/2022]
Abstract
G protein-coupled receptors (GPCRs) have important functions in both innate and adaptive immunity, with the capacity to bridge interactions between the two arms of the host responses to pathogens through direct recognition of secreted microbial products or the by-products of host cells damaged by pathogen exposure. In the mid-1990s, a large group of intracellular proteins was discovered, the regulator of G protein signaling (RGS) family, whose main, but not exclusive, function appears to be to constrain the intensity and duration of GPCR signaling. The R4/B subfamily--the focus of this review--includes RGS1-5, 8, 13, 16, 18, and 21, which are the smallest RGS proteins in size, with the exception of RGS3. Prominent roles in the trafficking of B and T lymphocytes and macrophages have been described for RGS1, RGS13, and RGS16, while RGS18 appears to control platelet and osteoclast functions. Additional G protein independent functions of RGS13 have been uncovered in gene expression in B lymphocytes and mast cell-mediated allergic reactions. In this review, we discuss potential physiological roles of this RGS protein subfamily, primarily in leukocytes having central roles in immune and inflammatory responses. We also discuss approaches to target RGS proteins therapeutically, which represents a virtually untapped strategy to combat exaggerated immune responses leading to inflammation.
Collapse
Affiliation(s)
- Zhihui Xie
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA
| | - Eunice C Chan
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA
| | - Kirk M Druey
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
49
|
Boularan C, Hwang IY, Kamenyeva O, Park C, Harrison K, Huang Z, Kehrl JH. B Lymphocyte-Specific Loss of Ric-8A Results in a Gα Protein Deficit and Severe Humoral Immunodeficiency. THE JOURNAL OF IMMUNOLOGY 2015; 195:2090-102. [PMID: 26232433 DOI: 10.4049/jimmunol.1500523] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/06/2015] [Indexed: 01/05/2023]
Abstract
Resistance to inhibitors of cholinesterase 8A (Ric-8A) is a highly evolutionarily conserved cytosolic protein initially identified in Caenorhabditis elegans, where it was assigned a regulatory role in asymmetric cell divisions. It functions as a guanine nucleotide exchange factor for Gαi, Gαq, and Gα12/13 and as a molecular chaperone required for the initial association of nascent Gα subunits with cellular membranes in embryonic stem cell lines. To test its role in hematopoiesis and B lymphocytes specifically, we generated ric8 (fl/fl) vav1-cre and ric8 (fl/fl) mb1-cre mice. The major hematopoietic cell lineages developed in the ric8 (fl/fl) vav1-cre mice, notwithstanding severe reduction in Gαi2/3, Gαq, and Gα13 proteins. B lymphocyte-specific loss of Ric-8A did not compromise bone marrow B lymphopoiesis, but splenic marginal zone B cell development failed, and B cells underpopulated lymphoid organs. The ric8 (fl/fl) mb1-cre B cells exhibited poor responses to chemokines, abnormal trafficking, improper in situ positioning, and loss of polarity components during B cell differentiation. The ric8 (fl/fl) mb1-cre mice had a severely disrupted lymphoid architecture and poor primary and secondary Ab responses. In B lymphocytes, Ric-8A is essential for normal Gα protein levels and is required for B cell differentiation, trafficking, and Ab responses.
Collapse
Affiliation(s)
- Cedric Boularan
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Il-Young Hwang
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Olena Kamenyeva
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Chung Park
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kathleen Harrison
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Zhen Huang
- Department of Neurology, University of Wisconsin-Madison, Madison, WI 53706; and Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53706
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
50
|
Neubig RR. RGS-Insensitive G Proteins as In Vivo Probes of RGS Function. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 133:13-30. [PMID: 26123300 DOI: 10.1016/bs.pmbts.2015.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Guanine nucleotide-binding proteins of the inhibitory (Gi/o) class play critical physiological roles and the receptors that activate them are important therapeutic targets (e.g., mu opioid, serotonin 5HT1a, etc.). Gi/o proteins are negatively regulated by regulator of G protein signaling (RGS) proteins. The redundant actions of the 20 different RGS family members have made it difficult to establish their overall physiological role. A unique G protein mutation (G184S in Gαi/o) prevents RGS binding to the Gα subunit and blocks all RGS action at that particular Gα subunit. The robust phenotypes of mice expressing these RGS-insensitive (RGSi) mutant G proteins illustrate the profound action of RGS proteins in cardiovascular, metabolic, and central nervous system functions. Specifically, the enhanced Gαi2 signaling through the RGSi Gαi2(G184S) mutant knock-in mice shows protection against cardiac ischemia/reperfusion injury and potentiation of serotonin-mediated antidepressant actions. In contrast, the RGSi Gαo mutant knock-in produces enhanced mu-opioid receptor-mediated analgesia but also a seizure phenotype. These genetic models provide novel insights into potential therapeutic strategies related to RGS protein inhibitors and/or G protein subtype-biased agonists at particular GPCRs.
Collapse
Affiliation(s)
- Richard R Neubig
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|