1
|
Kwak-Kim J, Maier CC, Villano CM, Bowman CJ, Brennan FR, Stanislaus D, Hillegas A, Krayer J, Prell RA, Papenfuss TL, Cauvin A, Gamse J, Dahlman A, Enright B, Leshin L, Rao GK, Helms W, Fuller CL, Yang X, Chen C, Mitchell-Ryan S. Assessing the impact and risk of immunomodulatory compounds on pregnancy. J Reprod Immunol 2025; 169:104453. [PMID: 39999662 DOI: 10.1016/j.jri.2025.104453] [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: 10/16/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
There have been remarkable advancements in understanding the complex and dynamic immune biological processes engaged during all stages of pregnancy. Exquisite control of immune processes is critical to successful outcome in all stages of pregnancy from ovulation to birth. There are many immunomodulatory therapeutics that may offer beneficial treatment options for a variety of diseases (e.g., inflammation/autoimmunity, cancer) to patients that are or desire to become pregnant. It is important to understand the potential for these immunomodulatory therapeutics to alter the critical immune processes in pregnancy to inform clinical risk relative to successful pregnancy. The Health and Environmental Sciences Institute-Developmental and Reproductive Toxicology/Immuno-safety Technical Committee (HESI DART/ITC) conducted a survey on approaches to assess adverse pregnancy outcomes with immunomodulators. HESI DART/ITC also organized a workshop for an extended discussion on immune mechanisms during pregnancy, the adequacy of current tools/methodologies to identify concerns for potential pregnancy hazards from immunomodulatory therapies, ways to identify and address scientific gaps, and global regulatory considerations across various immunomodulatory modalities and indications. In this manuscript we summarize learnings from these efforts to characterize risk within this patient population, promote more informed treatment decisions, and enable safer pharmacological interventions during pregnancy.
Collapse
Affiliation(s)
- Joanne Kwak-Kim
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Vernon Hills, IL, USA
| | | | - Caren M Villano
- Boehringer Ingelheim, Nonclinical Drug Safety, Ridgefield, CT, USA.
| | | | - Frank R Brennan
- Novartis Institute of BioMedical Research, Preclinical Safety (PCS), Basel, Switzerland
| | | | | | - John Krayer
- Johnson and Johnson, Non-clinical Safety, Springhouse, PA, USA
| | - Rodney A Prell
- Genentech, Inc., Department of Safety Assessment, South San Francisco, CA, USA
| | | | - Annick Cauvin
- UCB Biopharma SRL, Nonclinical Safety Evaluation, Brussels, Belgium
| | - Joshua Gamse
- Genmab, Non-Clinical Safety & Toxicology, Plainsboro, NJ, USA
| | - Anna Dahlman
- Genmab, Non-Clinical Safety & Toxicology, Copenhagen, Denmark
| | - Brian Enright
- AbbVie Inc., Preclinical Safety, North Chicago, IL, USA
| | - Lawrence Leshin
- United States Food and Drug Administration, CDER-OND-OII-DRTM, Silver Spring, MD, USA
| | - Gautham K Rao
- Genentech, Inc., Department of Safety Assessment, South San Francisco, CA, USA
| | | | | | - Xiuhua Yang
- The First Hospital of China Medical University, Department of Obstetrics and Gynecology, Shenyang, Liaoning, PR China
| | - Connie Chen
- The Health and Environmental Sciences Institute, Washington, DC, USA
| | | |
Collapse
|
2
|
Horn S, Schmid M, Berest I, Piattini F, Zhang J, de Bock K, Devuyst O, Nlandu Khodo S, Kisielow J, Kopf M. IL-1 protects from fatal systemic candidiasis in mice by inhibiting oxidative phosphorylation and hypoxia. Nat Commun 2025; 16:2626. [PMID: 40097388 PMCID: PMC11914259 DOI: 10.1038/s41467-025-57797-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Invasive C. albicans infections result in high mortality rates. While IL-1 is important to combat C. albicans infections, the underlying mechanisms remain unclear. Using global and conditional Il1r1 knockouts in mice, here we show that IL-1R signaling in non-hematopoietic cells in the kidney and brain is crucial for a protective response. In the kidney, endothelial IL-1R contributes to fungal clearance independent of neutrophil recruitment, while IL-1R in hematopoietic cells is dispensable. IL-1R signaling indirectly recruits neutrophils and monocytes in the brain by regulating chemokines and adhesion molecules. Single-nucleus-RNA-sequencing data implicates excessive metabolic activity and oxidative phosphorylation across all cell types in the kidney of Il1r1-deficient mice within a few hours upon infection, with associated, localized hypoxia at infection foci. Lastly, we find that hypoxia promotes fungal growth and pathogenicity. In summary, our results show that IL-1R-signaling in non-hematopoietic cells is required to prevent fatal candidiasis by inhibiting a metabolic shift, including excessive oxidative phosphorylation and hypoxia.
Collapse
Affiliation(s)
- Sofia Horn
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Mareike Schmid
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Ivan Berest
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Federica Piattini
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Jing Zhang
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zurich, Zurich, Switzerland
| | - Katrien de Bock
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zurich, Zurich, Switzerland
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Jan Kisielow
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Baker PJ, Bohrer AC, Castro E, Amaral EP, Snow-Smith M, Torres-Juárez F, Gould ST, Queiroz ATL, Fukutani ER, Jordan CM, Khillan JS, Cho K, Barber DL, Andrade BB, Johnson RF, Hilligan KL, Mayer-Barber KD. The inflammatory microenvironment of the lung at the time of infection governs innate control of SARS-CoV-2 replication. Sci Immunol 2024; 9:eadp7951. [PMID: 39642242 DOI: 10.1126/sciimmunol.adp7951] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/08/2024] [Indexed: 12/08/2024]
Abstract
Severity of COVID-19 is affected by multiple factors; however, it is not understood how the inflammatory milieu of the lung at the time of SARS-CoV-2 exposure affects the control of viral replication. Here, we demonstrate that immune events in the mouse lung closely preceding SARS-CoV-2 infection affect viral control and identify innate immune pathways that limit viral replication. Pulmonary inflammatory stimuli including resolved, antecedent respiratory infections with Staphylococcus aureus or influenza, ongoing pulmonary Mycobacterium tuberculosis infection, ovalbumin/alum-induced asthma, or airway administration of TLR ligands and recombinant cytokines all establish an antiviral state in the lung that restricts SARS-CoV-2 replication. In addition to antiviral type I interferons, TNFα and IL-1 potently precondition the lung for enhanced viral control. Our work shows that SARS-CoV-2 may benefit from an immunologically quiescent lung microenvironment and suggests that heterogeneity in pulmonary inflammation preceding SARS-CoV-2 exposure may contribute to variability in disease outcomes.
Collapse
Affiliation(s)
- Paul J Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Andrea C Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Ehydel Castro
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Eduardo P Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Maryonne Snow-Smith
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
- Human Eosinophil Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Flor Torres-Juárez
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sydnee T Gould
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Artur T L Queiroz
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Eduardo R Fukutani
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Cassandra M Jordan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jaspal S Khillan
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, MD 20852, USA
| | - Kyoungin Cho
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, MD 20852, USA
| | - Daniel L Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Bruno B Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Reed F Johnson
- SCV2 Virology Core, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Kerry L Hilligan
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Chen K, Xu B, Qiu S, Long L, Zhao Q, Xu J, Wang H. Inhibition of phosphodiesterase 4 attenuates aquaporin 4 expression and astrocyte swelling following cerebral ischemia/reperfusion injury. Glia 2024; 72:1629-1645. [PMID: 38785370 DOI: 10.1002/glia.24572] [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: 08/02/2023] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
We have previously shown that phosphodiesterase 4 (PDE4) inhibition protects against neuronal injury in rats following middle cerebral artery occlusion/reperfusion (MCAO/R). However, the effects of PDE4 on brain edema and astrocyte swelling are unknown. In this study, we showed that inhibition of PDE4 by Roflumilast (Roflu) reduced brain edema and brain water content in rats subjected to MCAO/R. Roflu decreased the expression of aquaporin 4 (AQP4), while the levels of phosphorylated protein kinase B (Akt) and forkhead box O3a (FoxO3a) were increased. In addition, Roflu reduced cell volume and the expression of AQP4 in primary astrocytes undergoing oxygen and glucose deprivation/reoxygenation (OGD/R). Consistently, PDE4B knockdown showed similar effects as PDE4 inhibition; and PDE4B overexpression rescued the inhibitory role of PDE4B knockdown on AQP4 expression. We then found that the effects of Roflu on the expression of AQP4 and cell volume were blocked by the Akt inhibitor MK2206. Since neuroinflammation and astrocyte activation are the common events that are observed in stroke, we treated primary astrocytes with interleukin-1β (IL-1β). Astrocytes treated with IL-1β showed decreased AQP4 and phosphorylated Akt and FoxO3a. Roflu significantly reduced AQP4 expression, which was accompanied by increased phosphorylation of Akt and FoxO3a. Furthermore, overexpression of FoxO3a partly reversed the effect of Roflu on AQP4 expression. Our findings suggest that PDE4 inhibition limits ischemia-induced brain edema and astrocyte swelling via the Akt/FoxO3a/AQP4 pathway. PDE4 is a promising target for the intervention of brain edema after cerebral ischemia.
Collapse
Affiliation(s)
- Kechun Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Bingtian Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuqin Qiu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Lu Long
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Qian Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiangping Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
| | - Haitao Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
| |
Collapse
|
5
|
Țichil I, Mitre I, Zdrenghea MT, Bojan AS, Tomuleasa CI, Cenariu D. A Review of Key Regulators of Steady-State and Ineffective Erythropoiesis. J Clin Med 2024; 13:2585. [PMID: 38731114 PMCID: PMC11084473 DOI: 10.3390/jcm13092585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Erythropoiesis is initiated with the transformation of multipotent hematopoietic stem cells into committed erythroid progenitor cells in the erythroblastic islands of the bone marrow in adults. These cells undergo several stages of differentiation, including erythroblast formation, normoblast formation, and finally, the expulsion of the nucleus to form mature red blood cells. The erythropoietin (EPO) pathway, which is activated by hypoxia, induces stimulation of the erythroid progenitor cells and the promotion of their proliferation and survival as well as maturation and hemoglobin synthesis. The regulation of erythropoiesis is a complex and dynamic interaction of a myriad of factors, such as transcription factors (GATA-1, STAT5), cytokines (IL-3, IL-6, IL-11), iron metabolism and cell cycle regulators. Multiple microRNAs are involved in erythropoiesis, mediating cell growth and development, regulating oxidative stress, erythrocyte maturation and differentiation, hemoglobin synthesis, transferrin function and iron homeostasis. This review aims to explore the physiology of steady-state erythropoiesis and to outline key mechanisms involved in ineffective erythropoiesis linked to anemia, chronic inflammation, stress, and hematological malignancies. Studying aberrations in erythropoiesis in various diseases allows a more in-depth understanding of the heterogeneity within erythroid populations and the development of gene therapies to treat hematological disorders.
Collapse
Affiliation(s)
- Ioana Țichil
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Ileana Mitre
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
| | - Mihnea Tudor Zdrenghea
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Anca Simona Bojan
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Ciprian Ionuț Tomuleasa
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
- MEDFUTURE—Research Centre for Advanced Medicine, 8 Louis Pasteur Street, 400347 Cluj-Napoca, Romania
| | - Diana Cenariu
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- MEDFUTURE—Research Centre for Advanced Medicine, 8 Louis Pasteur Street, 400347 Cluj-Napoca, Romania
| |
Collapse
|
6
|
Krishnamohan M, Kaplanov I, Maudi-Boker S, Yousef M, Machluf-Katz N, Cohen I, Elkabets M, Titus J, Bersudsky M, Apte RN, Voronov E, Braiman A. Tumor Cell-Associated IL-1α Affects Breast Cancer Progression and Metastasis in Mice through Manipulation of the Tumor Immune Microenvironment. Int J Mol Sci 2024; 25:3950. [PMID: 38612760 PMCID: PMC11011794 DOI: 10.3390/ijms25073950] [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: 02/25/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
IL-1α is a dual function cytokine that affects inflammatory and immune responses and plays a pivotal role in cancer. The effects of intracellular IL-1α on the development of triple negative breast cancer (TNBC) in mice were assessed using the CRISPR/Cas9 system to suppress IL-1α expression in 4T1 breast cancer cells. Knockout of IL-1α in 4T1 cells modified expression of multiple genes, including downregulation of cytokines and chemokines involved in the recruitment of tumor-associated pro-inflammatory cells. Orthotopical injection of IL-1α knockout (KO) 4T1 cells into BALB/c mice led to a significant decrease in local tumor growth and lung metastases, compared to injection of wild-type 4T1 (4T1/WT) cells. Neutrophils and myeloid-derived suppressor cells were abundant in tumors developing after injection of 4T1/WT cells, whereas more antigen-presenting cells were observed in the tumor microenvironment after injection of IL-1α KO 4T1 cells. This switch correlated with increased infiltration of CD3+CD8+ and NKp46+cells. Engraftment of IL-1α knockout 4T1 cells into immunodeficient NOD.SCID mice resulted in more rapid tumor growth, with increased lung metastasis in comparison to engraftment of 4T1/WT cells. Our results suggest that tumor-associated IL-1α is involved in TNBC progression in mice by modulating the interplay between immunosuppressive pro-inflammatory cells vs. antigen-presenting and cytotoxic cells.
Collapse
Affiliation(s)
- Mathumathi Krishnamohan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Irena Kaplanov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Sapir Maudi-Boker
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Muhammad Yousef
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Noy Machluf-Katz
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Idan Cohen
- Cancer Center, Emek Medical Center, Afula 18101, Israel;
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Jaison Titus
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Marina Bersudsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Ron N. Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| |
Collapse
|
7
|
Baker PJ, Bohrer AC, Castro E, Amaral EP, Snow-Smith M, Torres-Juárez F, Gould ST, Queiroz ATL, Fukutani ER, Jordan CM, Khillan JS, Cho K, Barber DL, Andrade BB, Johnson RF, Hilligan KL, Mayer-Barber KD. The inflammatory microenvironment of the lung at the time of infection governs innate control of SARS-CoV-2 replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586885. [PMID: 38585846 PMCID: PMC10996686 DOI: 10.1101/2024.03.27.586885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
SARS-CoV-2 infection leads to vastly divergent clinical outcomes ranging from asymptomatic infection to fatal disease. Co-morbidities, sex, age, host genetics and vaccine status are known to affect disease severity. Yet, how the inflammatory milieu of the lung at the time of SARS-CoV-2 exposure impacts the control of viral replication remains poorly understood. We demonstrate here that immune events in the mouse lung closely preceding SARS-CoV-2 infection significantly impact viral control and we identify key innate immune pathways required to limit viral replication. A diverse set of pulmonary inflammatory stimuli, including resolved antecedent respiratory infections with S. aureus or influenza, ongoing pulmonary M. tuberculosis infection, ovalbumin/alum-induced asthma or airway administration of defined TLR ligands and recombinant cytokines, all establish an antiviral state in the lung that restricts SARS-CoV-2 replication upon infection. In addition to antiviral type I interferons, the broadly inducible inflammatory cytokines TNFα and IL-1 precondition the lung for enhanced viral control. Collectively, our work shows that SARS-CoV-2 may benefit from an immunologically quiescent lung microenvironment and suggests that heterogeneity in pulmonary inflammation that precedes or accompanies SARS-CoV-2 exposure may be a significant factor contributing to the population-wide variability in COVID-19 disease outcomes.
Collapse
Affiliation(s)
- Paul J. Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- Current Address: Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Ehydel Castro
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Eduardo P. Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Maryonne Snow-Smith
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- Human Eosinophil Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Flor Torres-Juárez
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Sydnee T. Gould
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
- Current Address: Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Artur T. L. Queiroz
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Eduardo R. Fukutani
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Cassandra M. Jordan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Jaspal S. Khillan
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, Maryland 20852, USA
| | - Kyoungin Cho
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, Maryland 20852, USA
| | - Daniel L. Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Bruno B. Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Reed F. Johnson
- SCV2 Virology Core, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Kerry L. Hilligan
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| |
Collapse
|
8
|
Gölöncsér F, Baranyi M, Tod P, Maácz F, Sperlágh B. P2X7 receptor inhibition alleviates mania-like behavior independently of interleukin-1β. iScience 2024; 27:109284. [PMID: 38444608 PMCID: PMC10914489 DOI: 10.1016/j.isci.2024.109284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/15/2023] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Purinergic dysfunctions are associated with mania and depression pathogenesis. P2X7 receptor (P2X7R) mediates the IL-1β maturation via NLRP3 inflammasome activation. We tested in a mouse model of the subchronic amphetamine (AMPH)-induced hyperactivity whether P2X7R inhibition alleviated mania-like behavior through IL-1β. Treatment with JNJ-47965567, a P2X7R antagonist, abolished AMPH-induced hyperlocomotion in wild-type and IL-1α/β-knockout male mice. The NLRP3 inhibitor MCC950 failed to reduce AMPH-induced locomotion in WT mice, whereas the IL-1 receptor antagonist anakinra slightly increased it. AMPH increased IL-10, TNF-α, and TBARS levels, but did not influence BDNF levels, serotonin, dopamine, and noradrenaline content in brain tissues in either genotypes. JNJ-47965567 and P2rx7-gene deficiency, but not IL-1α/β-gene deficiency, attenuated AMPH-induced [3H]dopamine release from striatal slices. In wild-type and IL-1α/β-knockout female mice, JNJ-47965567 was also effective in attenuating AMPH-induced hyperlocomotion. This study suggests that AMPH-induced hyperactivity is modulated by P2X7Rs, but not through IL-1β.
Collapse
Affiliation(s)
- Flóra Gölöncsér
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Mária Baranyi
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Pál Tod
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Fruzsina Maácz
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University School of Ph.D Studies, 1083 Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University School of Ph.D Studies, 1083 Budapest, Hungary
| |
Collapse
|
9
|
Rai S, Zhang Y, Grockowiak E, Kimmerlin Q, Hansen N, Stoll CB, Usart M, Luque Paz D, Hao-Shen H, Zhu Y, Roux J, Bader MS, Dirnhofer S, Farady CJ, Schroeder T, Méndez-Ferrer S, Skoda RC. IL-1β promotes MPN disease initiation by favoring early clonal expansion of JAK2-mutant hematopoietic stem cells. Blood Adv 2024; 8:1234-1249. [PMID: 38207211 PMCID: PMC10912850 DOI: 10.1182/bloodadvances.2023011338] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024] Open
Abstract
ABSTRACT JAK 2-V617F is the most frequent somatic mutation causing myeloproliferative neoplasm (MPN). JAK2-V617F can be found in healthy individuals with clonal hematopoiesis of indeterminate potential (CHIP) with a frequency much higher than the prevalence of MPNs. The factors controlling the conversion of JAK2-V617F CHIP to MPN are largely unknown. We hypothesized that interleukin-1β (IL-1β)-mediated inflammation can favor this progression. We established an experimental system using bone marrow (BM) transplantations from JAK2-V617F and GFP transgenic (VF;GFP) mice that were further crossed with IL-1β-/- or IL-1R1-/- mice. To study the role of IL-1β and its receptor on monoclonal evolution of MPN, we performed competitive BM transplantations at high dilutions with only 1 to 3 hematopoietic stem cells (HSCs) per recipient. Loss of IL-1β in JAK2-mutant HSCs reduced engraftment, restricted clonal expansion, lowered the total numbers of functional HSCs, and decreased the rate of conversion to MPN. Loss of IL-1R1 in the recipients also lowered the conversion to MPN but did not reduce the frequency of engraftment of JAK2-mutant HSCs. Wild-type (WT) recipients transplanted with VF;GFP BM that developed MPNs had elevated IL-1β levels and reduced frequencies of mesenchymal stromal cells (MSCs). Interestingly, frequencies of MSCs were also reduced in recipients that did not develop MPNs, had only marginally elevated IL-1β levels, and displayed low GFP-chimerism resembling CHIP. Anti-IL-1β antibody preserved high frequencies of MSCs in VF;GFP recipients and reduced the rate of engraftment and the conversion to MPN. Our results identify IL-1β as a potential therapeutic target for preventing the transition from JAK2-V617F CHIP to MPNs.
Collapse
Affiliation(s)
- Shivam Rai
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Yang Zhang
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Elodie Grockowiak
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Quentin Kimmerlin
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nils Hansen
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Cedric B. Stoll
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marc Usart
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Damien Luque Paz
- University of Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
| | - Hui Hao-Shen
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Yexuan Zhu
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Julien Roux
- Department of Biomedicine, Bioinformatics core facility, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Michael S. Bader
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| | - Stefan Dirnhofer
- Department of Pathology, University Hospital Basel, Basel, Switzerland
| | | | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Simón Méndez-Ferrer
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Radek C. Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
10
|
Matsuda R, Sorobetea D, Zhang J, Peterson ST, Grayczyk JP, Yost W, Apenes N, Kovalik ME, Herrmann B, O’Neill RJ, Bohrer AC, Lanza M, Assenmacher CA, Mayer-Barber KD, Shin S, Brodsky IE. A TNF-IL-1 circuit controls Yersinia within intestinal pyogranulomas. J Exp Med 2024; 221:e20230679. [PMID: 38363547 PMCID: PMC10873131 DOI: 10.1084/jem.20230679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/22/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024] Open
Abstract
Tumor necrosis factor (TNF) is a pleiotropic inflammatory cytokine that mediates antimicrobial defense and granuloma formation in response to infection by numerous pathogens. We previously reported that Yersinia pseudotuberculosis colonizes the intestinal mucosa and induces the recruitment of neutrophils and inflammatory monocytes into organized immune structures termed pyogranulomas (PG) that control Yersinia infection. Inflammatory monocytes are essential for the control and clearance of Yersinia within intestinal PG, but how monocytes mediate Yersinia restriction is poorly understood. Here, we demonstrate that TNF signaling in monocytes is required for bacterial containment following enteric Yersinia infection. We further show that monocyte-intrinsic TNFR1 signaling drives the production of monocyte-derived interleukin-1 (IL-1), which signals through IL-1 receptors on non-hematopoietic cells to enable PG-mediated control of intestinal Yersinia infection. Altogether, our work reveals a monocyte-intrinsic TNF-IL-1 collaborative inflammatory circuit that restricts intestinal Yersinia infection.
Collapse
Affiliation(s)
- Rina Matsuda
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Sorobetea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jenna Zhang
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefan T. Peterson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James P. Grayczyk
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Winslow Yost
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolai Apenes
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria E. Kovalik
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Beatrice Herrmann
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rosemary J. O’Neill
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew Lanza
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Igor E. Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Martínez-Ramos S, García S. An update of murine models and their methodologies in immune-mediated joint damage and pain research. Int Immunopharmacol 2024; 128:111440. [PMID: 38176343 DOI: 10.1016/j.intimp.2023.111440] [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: 11/15/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Murine models have played an indispensable role in the understanding of rheumatic and musculoskeletal disorders (RMD), elucidating the genetic, endocrine and biomechanical pathways involved in joint pathology and associated pain. To date, the available models in RMD can be classified as induced or spontaneous, both incorporating transgenic alternatives that improve specific insights. It is worth noting that the selection of the most appropriate model together with the evaluation of their specific characteristics and technical capabilities are crucial when designing the experiments. Furthermore, it is also imperative to consistently adhere to the ethical standards concerning animal experimentation. Recognizing the inherent limitation that any model can entirely encapsulates the complexity of the pathophysiology of these conditions, the aim of this review is to provide an updated overview on the methodology of current murine models in major arthropathies and their immune-mediated pathways, addressing to basic, translational and pharmacological research in joint damage and pain.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain.
| | - Samuel García
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| |
Collapse
|
12
|
Pyrillou K, Humphry M, Kitt LA, Rodgers A, Nus M, Bennett MR, Smith KG, Lyons PA, Mallat Z, Clarke MC. Loss of T follicular regulatory cell-derived IL-1R2 augments germinal center reactions via increased IL-1. JCI Insight 2024; 9:e174005. [PMID: 38329807 PMCID: PMC11143922 DOI: 10.1172/jci.insight.174005] [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: 07/18/2023] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Inappropriate immune activity is key in the pathogenesis of multiple diseases, and it is typically driven by excess inflammation and/or autoimmunity. IL-1 is often the effector owing to its powerful role in both innate and adaptive immunity, and, thus, it is tightly controlled at multiple levels. IL-1R2 antagonizes IL-1, but effects of losing this regulation are unknown. We found that IL-1R2 resolves inflammation by rapidly scavenging free IL-1. Specific IL-1R2 loss in germinal center (GC) T follicular regulatory (Tfr) cells increased the GC response after a first, but not booster, immunization, with an increase in T follicular helper (Tfh) cells, GC B cells, and antigen-specific antibodies, which was reversed upon IL-1 blockade. However, IL-1 signaling is not obligate for GC reactions, as WT and Il1r1-/- mice showed equivalent phenotypes, suggesting that GC IL-1 is normally restrained by IL-1R2. Fascinatingly, germline Il1r2-/- mice did not show this phenotype, but conditional Il1r2 deletion in adulthood recapitulated it, implying that compensation during development counteracts IL-1R2 loss. Finally, patients with ulcerative colitis or Crohn's disease had lower serum IL-1R2. All together, we show that IL-1R2 controls important aspects of innate and adaptive immunity and that IL-1R2 level may contribute to human disease propensity and/or progression.
Collapse
Affiliation(s)
- Katerina Pyrillou
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Melanie Humphry
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Lauren A. Kitt
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Amanda Rodgers
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Meritxell Nus
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Martin R. Bennett
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Kenneth G.C. Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Paul A. Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Ziad Mallat
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Murray C.H. Clarke
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| |
Collapse
|
13
|
Fergany A, Zong C, Ekuban FA, Wu B, Ueha S, Shichino S, Matsushima K, Iwakura Y, Ichihara S, Ichihara G. Transcriptome analysis of the cerebral cortex of acrylamide-exposed wild-type and IL-1β-knockout mice. Arch Toxicol 2024; 98:181-205. [PMID: 37971544 PMCID: PMC10761544 DOI: 10.1007/s00204-023-03627-9] [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: 09/09/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Acrylamide is an environmental electrophile that has been produced in large amounts for many years. There is concern about the adverse health effects of acrylamide exposure due to its widespread industrial use and also presence in commonly consumed foods and others. IL-1β is a key cytokine that protects the brain from inflammatory insults, but its role in acrylamide-induced neurotoxicity remains unknown. We reported recently that deletion of IL-1β gene exacerbates ACR-induced neurotoxicity in mice. The aim of this study was to identify genes or signaling pathway(s) involved in enhancement of ACR-induced neurotoxicity by IL-1β gene deletion or ACR-induced neurotoxicity to generate a hypothesis mechanism explaining ACR-induced neurotoxicity. C57BL/6 J wild-type and IL-1β KO mice were exposed to ACR at 0, 12.5, 25 mg/kg by oral gavage for 7 days/week for 4 weeks, followed by extraction of mRNA from mice cerebral cortex for RNA sequence analysis. IL-1β deletion altered the expression of genes involved in extracellular region, including upregulation of PFN1 gene related to amyotrophic lateral sclerosis and increased the expression of the opposite strand of IL-1β. Acrylamide exposure enhanced mitochondria oxidative phosphorylation, synapse and ribosome pathways, and activated various pathways of different neurodegenerative diseases, such as Alzheimer disease, Parkinson disease, Huntington disease, and prion disease. Protein network analysis suggested the involvement of different proteins in related to learning and cognitive function, such as Egr1, Egr2, Fos, Nr4a1, and Btg2. Our results identified possible pathways involved in IL-1β deletion-potentiated and ACR-induced neurotoxicity in mice.
Collapse
Affiliation(s)
- Alzahraa Fergany
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
- Laboratory of Genetics and Genetic Engineering in Department of Animal Husbandry and Animal Wealth Development, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Cai Zong
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Frederick Adams Ekuban
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Bin Wu
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yoichiro Iwakura
- Division of Experimental Animal Immunology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
14
|
Wang Y, Morishima T, Sezaki M, Sato R, Nakato G, Fukuda S, Kobiyama K, Ishii KJ, Li Y, Takizawa H. Akkermansia muciniphila induces slow extramedullary hematopoiesis via cooperative IL-1R/TLR signals. EMBO Rep 2023; 24:e57485. [PMID: 37870318 PMCID: PMC10702838 DOI: 10.15252/embr.202357485] [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: 05/14/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
Bacterial infections can activate and mobilize hematopoietic stem and progenitor cells (HSPCs) from the bone marrow (BM) to the spleen, a process termed extramedullary hematopoiesis (EMH). Recent studies suggest that commensal bacteria regulate not only the host immune system but also hematopoietic homeostasis. However, the impact of gut microbes on hematopoietic pathology remains unclear. Here, we find that systemic single injections of Akkermansia muciniphila (A. m.), a mucin-degrading bacterium, rapidly activate BM myelopoiesis and slow but long-lasting hepato-splenomegaly, characterized by the expansion and differentiation of functional HSPCs, which we term delayed EMH. Mechanistically, delayed EMH triggered by A. m. is mediated entirely by the MYD88/TRIF innate immune signaling pathway, which persistently stimulates splenic myeloid cells to secrete interleukin (IL)-1α, and in turn, activates IL-1 receptor (IL-1R)-expressing splenic HSPCs. Genetic deletion of Toll-like receptor-2 and -4 (TLR2/4) or IL-1α partially diminishes A. m.-induced delayed EMH, while inhibition of both pathways alleviates splenomegaly and EMH. Our results demonstrate that cooperative IL-1R- and TLR-mediated signals regulate commensal bacteria-driven EMH, which might be relevant for certain autoimmune disorders.
Collapse
Affiliation(s)
- Yuxin Wang
- Department of Hematology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Tatsuya Morishima
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
- Laboratory of Hematopoietic Stem Cell Engineering, IRCMSKumamoto UniversityKumamotoJapan
| | - Maiko Sezaki
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
- Laboratory of Hematopoietic Stem Cell Engineering, IRCMSKumamoto UniversityKumamotoJapan
| | - Ryo Sato
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Gaku Nakato
- Gut Environmental Design GroupKanagawa Institute of Industrial Science and TechnologyAtsugiJapan
| | - Shinji Fukuda
- Gut Environmental Design GroupKanagawa Institute of Industrial Science and TechnologyAtsugiJapan
- Institute for Advanced Biosciences (IAB)Keio UniversityTokyoJapan
- Transborder Medical Research CenterUniversity of TsukubaTsukubaJapan
| | - Kouji Kobiyama
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical ScienceThe University of TokyoTokyoJapan
- International Vaccine Design Center, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Ken J Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical ScienceThe University of TokyoTokyoJapan
- International Vaccine Design Center, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Yuhua Li
- Department of Hematology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA)Kumamoto UniversityKumamotoJapan
| |
Collapse
|
15
|
Griffith JW, Faustino LD, Cottrell VI, Nepal K, Hariri LP, Chiu RSY, Jones MC, Julé A, Gabay C, Luster AD. Regulatory T cell-derived IL-1Ra suppresses the innate response to respiratory viral infection. Nat Immunol 2023; 24:2091-2107. [PMID: 37945820 PMCID: PMC11887468 DOI: 10.1038/s41590-023-01655-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 09/15/2023] [Indexed: 11/12/2023]
Abstract
Regulatory T (Treg) cell modulation of adaptive immunity and tissue homeostasis is well described; however, less is known about Treg cell-mediated regulation of the innate immune response. Here we show that deletion of ST2, the receptor for interleukin (IL)-33, on Treg cells increased granulocyte influx into the lung and increased cytokine production by innate lymphoid and γδ T cells without alteration of adaptive immunity to influenza. IL-33 induced high levels of the interleukin-1 receptor antagonist (IL-1Ra) in ST2+ Treg cells and deletion of IL-1Ra in Treg cells increased granulocyte influx into the lung. Treg cell-specific deletion of ST2 or IL-1Ra improved survival to influenza, which was dependent on IL-1. Adventitial fibroblasts in the lung expressed high levels of the IL-1 receptor and their chemokine production was suppressed by Treg cell-produced IL-1Ra. Thus, we define a new pathway where IL-33-induced IL-1Ra production by tissue Treg cells suppresses IL-1-mediated innate immune responses to respiratory viral infection.
Collapse
Affiliation(s)
- Jason W Griffith
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lucas D Faustino
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Victoria I Cottrell
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keshav Nepal
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lida P Hariri
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca Suet-Yan Chiu
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael C Jones
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amélie Julé
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Cem Gabay
- Division of Rheumatology, University Hospitals of Geneva and University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Suzuki T, Boonyaleka K, Okano T, Iida T, Yoshida M, Fukano H, Hoshino Y, Iwakura Y, Ablordey AS, Ashida H. Inflammasome-triggered IL-18 controls skin inflammation in the progression of Buruli ulcer. PLoS Pathog 2023; 19:e1011747. [PMID: 37910490 PMCID: PMC10619818 DOI: 10.1371/journal.ppat.1011747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023] Open
Abstract
Buruli ulcer is an emerging chronic infectious skin disease caused by Mycobacterium ulcerans. Mycolactone, an exotoxin produced by the bacterium, is the only identified virulence factor so far, but the functions of this toxin and the mechanisms of disease progression remain unclear. By interfering Sec61 translocon, mycolactone inhibits the Sec61-dependent co-translational translocation of newly synthesized proteins, such as induced cytokines and immune cell receptors, into the endoplasmic reticulum. However, in regard to IL-1β, which is secreted by a Sec61-independent mechanism, mycolactone has been shown to induce IL-1β secretion via activation of inflammasomes. In this study, we clarified that cytokine induction, including that of IL-1β, in infected macrophages was suppressed by mycolactone produced by M. ulcerans subsp. shinshuense, despite the activation of caspase-1 through the inflammasome activation triggered in a manner independent of mycolactone. Intriguingly, mycolactone suppressed the expression of proIL-1β as well as TNF-α at the transcriptional level, suggesting that mycolactone of M. ulcerans subsp. shinshuense may exert additional inhibitory effect on proIL-1β expression. Remarkably, constitutively produced IL-18 was cleaved and mature IL-18 was actually released from macrophages infected with the causative mycobacterium. IL-18-deficient mice infected subcutaneously with M. ulcerans exhibited exacerbated skin inflammation during the course of disease progression. On the other hand, IL-1β controls bacterial multiplication in skin tissues. These results provide information regarding the mechanisms and functions of the induced cytokines in the pathology of Buruli ulcer.
Collapse
Affiliation(s)
- Toshihiko Suzuki
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kotchakorn Boonyaleka
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tokuju Okano
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tamako Iida
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Mitsunori Yoshida
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hanako Fukano
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshihiko Hoshino
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Anthony S. Ablordey
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Hiroshi Ashida
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
17
|
Maeder C, Speer T, Wirth A, Boeckel JN, Fatima S, Shahzad K, Freichel M, Laufs U, Gaul S. Membrane-bound Interleukin-1α mediates leukocyte adhesion during atherogenesis. Front Immunol 2023; 14:1252384. [PMID: 37701434 PMCID: PMC10494239 DOI: 10.3389/fimmu.2023.1252384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/10/2023] [Indexed: 09/14/2023] Open
Abstract
Introduction The interleukin-1 (IL-1) family and the NLR family pyrin domain-containing 3 (NLRP3) inflammasome contribute to atherogenesis but the underlying mechanisms are incompletely understood. Unlike IL-1β, IL-1α is not dependent on the NLRP3 inflammasome to exert its pro-inflammatory effects. Here, a non-genetic model was applied to characterize the role of IL-1α, IL-1β, and NLRP3 for the pathogenesis of atherosclerosis. Methods Atherogenesis was induced by gain-of-function PCSK9-AAV8 mutant viruses and feeding of a high-fat western diet (WTD) for 12 weeks in C57Bl6/J wildtype mice (WT) and in Il1a-/-, Nlrp3-/-, and Il1b-/- mice. Results PCSK9-Il1a-/- mice showed reduced atherosclerotic plaque area in the aortic root with lower lipid accumulation, while no difference was observed between PCSK9-WT, PCSK9-Nlrp3-/- and PCSK9-Il1b-/- mice. Serum proteomic analysis showed a reduction of pro-inflammatory cytokines (e.g., IL-1β, IL-6) in PCSK9-Il1a-/- as well as in PCSK9-Nlrp3-/- and PCSK9-Il1b-/- mice. Bone marrow dendritic cells (BMDC) of PCSK9-WT, PCSK9-Nlrp3-/-, and PCSK9-Il1b-/- mice and primary human monocytes showed translocation of IL-1α to the plasma membrane (csIL-1α) upon stimulation with LPS. The translocation of IL-1α to the cell surface was regulated by myristoylation and increased in mice with hypercholesterolemia. CsIL-1α and IL1R1 protein-protein interaction on endothelial cells induced VCAM1 expression and monocyte adhesion, which was abrogated by the administration of neutralizing antibodies against IL-1α and IL1R1. Conclusion The results highlight the importance of IL-1α on the cell surface of circulating leucocytes for the development of atherosclerosis. PCSK9-Il1a-/- mice, but not PCSK9-Nlrp3-/- or PCSK9-Il1b-/- mice, are protected from atherosclerosis after induction of hypercholesterolemia independent of circulating cytokines. Myristoylation and translocation of IL-1α to the cell surface in myeloid cells facilitates leukocyte adhesion and contributes to the development of atherosclerosis.
Collapse
Affiliation(s)
- Christina Maeder
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Leipzig, Germany
| | - Thimoteus Speer
- Medizinische Klinik 4, Nephrologie, Goethe-Universität Frankfurt, Frankfurt am Main, Germany
- Else Kroener Fresenius Zentrum für Nephrologische Forschung, Goethe-Universität Frankfurt, Frankfurt am Main, Germany
| | - Angela Wirth
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Jes-Niels Boeckel
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Leipzig, Germany
| | - Sameen Fatima
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, Universitätsklinikum Leipzig, Leipzig University, Leipzig, Germany
| | - Khurrum Shahzad
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, Universitätsklinikum Leipzig, Leipzig University, Leipzig, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Leipzig, Germany
| | - Susanne Gaul
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig University, Leipzig, Germany
| |
Collapse
|
18
|
Sattiraju A, Kang S, Giotti B, Chen Z, Marallano VJ, Brusco C, Ramakrishnan A, Shen L, Tsankov AM, Hambardzumyan D, Friedel RH, Zou H. Hypoxic niches attract and sequester tumor-associated macrophages and cytotoxic T cells and reprogram them for immunosuppression. Immunity 2023; 56:1825-1843.e6. [PMID: 37451265 PMCID: PMC10527169 DOI: 10.1016/j.immuni.2023.06.017] [Citation(s) in RCA: 109] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 02/24/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
Glioblastoma (GBM), a highly lethal brain cancer, is notorious for immunosuppression, but the mechanisms remain unclear. Here, we documented a temporospatial patterning of tumor-associated myeloid cells (TAMs) corresponding to vascular changes during GBM progression. As tumor vessels transitioned from the initial dense regular network to later scant and engorged vasculature, TAMs shifted away from perivascular regions and trafficked to vascular-poor areas. This process was heavily influenced by the immunocompetence state of the host. Utilizing a sensitive fluorescent UnaG reporter to track tumor hypoxia, coupled with single-cell transcriptomics, we revealed that hypoxic niches attracted and sequestered TAMs and cytotoxic T lymphocytes (CTLs), where they were reprogrammed toward an immunosuppressive state. Mechanistically, we identified chemokine CCL8 and cytokine IL-1β as two hypoxic-niche factors critical for TAM trafficking and co-evolution of hypoxic zones into pseudopalisading patterns. Therefore, perturbation of TAM patterning in hypoxic zones may improve tumor control.
Collapse
Affiliation(s)
- Anirudh Sattiraju
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sangjo Kang
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhihong Chen
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Valerie J Marallano
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Concetta Brusco
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander M Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dolores Hambardzumyan
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Roland H Friedel
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Hongyan Zou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
19
|
Kakutani K, Yurube T, An HS, Doita M, Masuda K. Cytokine Inhibitors Upregulate Extracellular Matrix Anabolism of Human Intervertebral Discs under Alginate Beads and Alginate-Embedded Explant Cultures. Int J Mol Sci 2023; 24:12336. [PMID: 37569715 PMCID: PMC10418414 DOI: 10.3390/ijms241512336] [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: 06/12/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
We investigated the effects of the cytokine inhibitors IL-1 receptor antagonist (IL-1Ra) and soluble tumor necrosis factor receptor-1 (sTNFR1) on the extracellular matrix metabolism of human intervertebral discs (IVDs) and the roles of IL-1β and TNF in the homeostasis of IVD cells. The 1.2% alginate beads and the explants obtained from 35 human lumbar discs were treated with cytokine inhibitors. Extracellular matrix metabolism was evaluated by proteoglycan (PG) and collagen syntheses and IL-1β, TNF, and IL-6 expressions after three days of culture in the presence or absence of IL-1Ra, sTNFR1, and cycloheximide. Simultaneous treatment with IL-1Ra and sTNFR1 stimulated PG and collagen syntheses in the NP and AF cells and explants. The IL-1β concentration was significantly correlated to the relative increase in PG synthesis in AF explants after simultaneous cytokine inhibitor treatment. The relative increase in PG synthesis induced by simultaneous cytokine treatment was significantly higher in an advanced grade of MRI. Expressions of IL-1β and TNF were upregulated by each cytokine inhibitor, and simultaneous treatment suppressed IL-1β and TNF productions. In conclusion, IL-1Ra and sTNFR1 have the potential to increase PG and collagen synthesis in IVDs. IL-1β and TNF have a feedback pathway to maintain optimal expression, resulting in the control of homeostasis in IVD explants.
Collapse
Affiliation(s)
- Kenichiro Kakutani
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan;
- Department of Orthopaedic Surgery, Rush University Medical Center, Orthopaedic Building, Suite 300, 1611 W Harrison Street, Chicago, IL 60612, USA;
| | - Takashi Yurube
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan;
| | - Howard S. An
- Department of Orthopaedic Surgery, Rush University Medical Center, Orthopaedic Building, Suite 300, 1611 W Harrison Street, Chicago, IL 60612, USA;
| | - Minoru Doita
- Department of Orthopedic Surgery, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba-cho, Showa-gun, Iwate 028-3895, Japan;
| | - Koichi Masuda
- Department of Orthopaedic Surgery, University of California, San Diego, 9500 Gilman Dr. Mail Code 0863, La Jolla, CA 92093-0863, USA;
| |
Collapse
|
20
|
Fujimura K, Karasawa T, Komada T, Yamada N, Mizushina Y, Baatarjav C, Matsumura T, Otsu K, Takeda N, Mizukami H, Kario K, Takahashi M. NLRP3 inflammasome-driven IL-1β and IL-18 contribute to lipopolysaccharide-induced septic cardiomyopathy. J Mol Cell Cardiol 2023; 180:58-68. [PMID: 37172930 DOI: 10.1016/j.yjmcc.2023.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
Sepsis is a life-threatening syndrome, and its associated mortality is increased when cardiac dysfunction and damage (septic cardiomyopathy [SCM]) occur. Although inflammation is involved in the pathophysiology of SCM, the mechanism of how inflammation induces SCM in vivo has remained obscure. NLRP3 inflammasome is a critical component of the innate immune system that activates caspase-1 (Casp1) and causes the maturation of IL-1β and IL-18 as well as the processing of gasdermin D (GSDMD). Here, we investigated the role of the NLRP3 inflammasome in a murine model of lipopolysaccharide (LPS)-induced SCM. LPS injection induced cardiac dysfunction, damage, and lethality, which was significantly prevented in NLRP3-/- mice, compared to wild-type (WT) mice. LPS injection upregulated mRNA levels of inflammatory cytokines (Il6, Tnfa, and Ifng) in the heart, liver, and spleen of WT mice, and this upregulation was prevented in NLRP3-/- mice. LPS injection increased plasma levels of inflammatory cytokines (IL-1β, IL-18, and TNF-α) in WT mice, and this increase was markedly inhibited in NLRP3-/- mice. LPS-induced SCM was also prevented in Casp1/11-/- mice, but not in Casp11mt, IL-1β-/-, IL-1α-/-, or GSDMD-/- mice. Notably, LPS-induced SCM was apparently prevented in IL-1β-/- mice transduced with adeno-associated virus vector expressing IL-18 binding protein (IL-18BP). Furthermore, splenectomy, irradiation, or macrophage depletion alleviated LPS-induced SCM. Our findings demonstrate that the cross-regulation of NLRP3 inflammasome-driven IL-1β and IL-18 contributes to the pathophysiology of SCM and provide new insights into the mechanism underlying the pathogenesis of SCM.
Collapse
Affiliation(s)
- Kenta Fujimura
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tadayoshi Karasawa
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Takanori Komada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Naoya Yamada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Yoshiko Mizushina
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Chintogtokh Baatarjav
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Takayoshi Matsumura
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Kinya Otsu
- The School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom; National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
21
|
Matsuda R, Sorobetea D, Zhang J, Peterson ST, Grayczyk JP, Herrmann B, Yost W, O’Neill R, Bohrer AC, Lanza M, Assenmacher CA, Mayer-Barber KD, Shin S, Brodsky IE. A TNF-IL-1 circuit controls Yersinia within intestinal granulomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537749. [PMID: 37197029 PMCID: PMC10176537 DOI: 10.1101/2023.04.21.537749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Tumor necrosis factor (TNF) is a pleiotropic inflammatory cytokine that mediates antimicrobial defense and granuloma formation in response to infection by numerous pathogens. Yersinia pseudotuberculosis colonizes the intestinal mucosa and induces recruitment of neutrophils and inflammatory monocytes into organized immune structures termed pyogranulomas that control the bacterial infection. Inflammatory monocytes are essential for control and clearance of Yersinia within intestinal pyogranulomas, but how monocytes mediate Yersinia restriction is poorly understood. Here, we demonstrate that TNF signaling in monocytes is required for bacterial containment following enteric Yersinia infection. We further show that monocyte-intrinsic TNFR1 signaling drives production of monocyte-derived interleukin-1 (IL-1), which signals through IL-1 receptor on non-hematopoietic cells to enable pyogranuloma-mediated control of Yersinia infection. Altogether, our work reveals a monocyte-intrinsic TNF-IL-1 collaborative circuit as a crucial driver of intestinal granuloma function, and defines the cellular target of TNF signaling that restricts intestinal Yersinia infection.
Collapse
Affiliation(s)
- Rina Matsuda
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Daniel Sorobetea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Jenna Zhang
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Stefan T. Peterson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - James P. Grayczyk
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Beatrice Herrmann
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Winslow Yost
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Rosemary O’Neill
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew Lanza
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Igor E. Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
22
|
Spella M, Ntaliarda G, Skiadas G, Lamort AS, Vreka M, Marazioti A, Lilis I, Bouloukou E, Giotopoulou GA, Pepe MAA, Weiss SAI, Petrera A, Hauck SM, Koch I, Lindner M, Hatz RA, Behr J, Arendt KAM, Giopanou I, Brunn D, Savai R, Jenne DE, de Château M, Yull FE, Blackwell TS, Stathopoulos GT. Non-Oncogene Addiction of KRAS-Mutant Cancers to IL-1β via Versican and Mononuclear IKKβ. Cancers (Basel) 2023; 15:1866. [PMID: 36980752 PMCID: PMC10047096 DOI: 10.3390/cancers15061866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Kirsten rat sarcoma virus (KRAS)-mutant cancers are frequent, metastatic, lethal, and largely undruggable. While interleukin (IL)-1β and nuclear factor (NF)-κB inhibition hold promise against cancer, untargeted treatments are not effective. Here, we show that human KRAS-mutant cancers are addicted to IL-1β via inflammatory versican signaling to macrophage inhibitor of NF-κB kinase (IKK) β. Human pan-cancer and experimental NF-κB reporter, transcriptome, and proteome screens reveal that KRAS-mutant tumors trigger macrophage IKKβ activation and IL-1β release via secretory versican. Tumor-specific versican silencing and macrophage-restricted IKKβ deletion prevents myeloid NF-κB activation and metastasis. Versican and IKKβ are mutually addicted and/or overexpressed in human cancers and possess diagnostic and prognostic power. Non-oncogene KRAS/IL-1β addiction is abolished by IL-1β and TLR1/2 inhibition, indicating cardinal and actionable roles for versican and IKKβ in metastasis.
Collapse
Affiliation(s)
- Magda Spella
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Giannoula Ntaliarda
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Georgios Skiadas
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Anne-Sophie Lamort
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Malamati Vreka
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Antonia Marazioti
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Ioannis Lilis
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Eleni Bouloukou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Georgia A. Giotopoulou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Mario A. A. Pepe
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Stefanie A. I. Weiss
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Agnese Petrera
- Research Unit Protein Science-Core Facility Proteomics, Helmholtz Center Munich–German Research Center for Environmental Health, 80939 Munich, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science-Core Facility Proteomics, Helmholtz Center Munich–German Research Center for Environmental Health, 80939 Munich, Germany
| | - Ina Koch
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Michael Lindner
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Rudolph A. Hatz
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Juergen Behr
- Department of Internal Medicine V, Ludwig-Maximilian-University of Munich, 81377 Munich, Germany
| | - Kristina A. M. Arendt
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Ioanna Giopanou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - David Brunn
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60596 Frankfurt am Main, Germany
- Department of Internal Medicine and Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Dieter E. Jenne
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
- Max-Planck-Institute of Neurobiology, 82152 Planegg, Germany
| | | | - Fiona E. Yull
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Timothy S. Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Georgios T. Stathopoulos
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
23
|
Ashino T, Nakamura Y, Ohtaki H, Iwakura Y, Numazawa S. Downregulation of the gene expression of Cyp2c29 and Cyp3a11 by cecal ligation and puncture-induced sepsis is associated with interleukin-6. Int Immunopharmacol 2023; 117:110039. [PMID: 36944277 DOI: 10.1016/j.intimp.2023.110039] [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: 01/29/2023] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/22/2023]
Abstract
Sepsis is a pathological condition that affects the metabolism of administered drugs, leading to changes in the duration and intensity of their intended efficacies. Proinflammatory cytokines downregulate the expression of cytochrome P450s (P450s). The effects of P450 expression under inflammatory conditions have been studied using prophlogistic substances such as lipopolysaccharide; however, few studies have focused on clinical models of sepsis. Here, we show that cecal ligation and puncture (CLP), an approach for the study of human polymicrobial sepsis, leads to the expression of interleukin-1β (IL-1β), IL-6, and tumor necrosis factor α (TNFα) at 24 h after the CLP operation. Following CLP, IL-6-/- mice exhibited markedly lower survival than WT mice. In addition, CLP led to the significant downregulation of Cyp2c29 and Cyp3a11 gene expression in IL-1α-/-/β-/- (IL-1-/-) and TNFα-/- mice as well as in WT mice. In contrast, CLP elicited no significant effect on Cyp3a11 expression in IL-6-/- mice. Although CLP reduced the Cyp2c29 expression level in IL-6-/- mice, the expression of Cyp2c29 was lower in CLP-operated WT mice than in CLP-operated IL-6-/- mice. The reduction in the respective P450 protein levels and activities due to CLP-induced sepsis, reflected in the mRNA expression levels, was abolished by IL-6 depletion. Thus, CLP-induced sepsis downregulates P450 gene expression, particularly Cyp2c expression, and this effect is associated with IL-6 without affecting resistance to CLP-induced sepsis. These findings demonstrate the usefulness of CLP for studying the regulation of P450s and highlight IL-6 as a potential indicator of drug-metabolizing capacity under septic conditions.
Collapse
Affiliation(s)
- Takashi Ashino
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Yuki Nakamura
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Center for Pharmaceutical Education, Faculty of Pharmacy, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama, Kanagawa 245-0066, Japan
| | - Hirokazu Ohtaki
- Department of Functional Neurobiology, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba 278-0022, Japan
| | - Satoshi Numazawa
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| |
Collapse
|
24
|
Fülöp B, Hunyady Á, Bencze N, Kormos V, Szentes N, Dénes Á, Lénárt N, Borbély É, Helyes Z. IL-1 Mediates Chronic Stress-Induced Hyperalgesia Accompanied by Microglia and Astroglia Morphological Changes in Pain-Related Brain Regions in Mice. Int J Mol Sci 2023; 24:ijms24065479. [PMID: 36982563 PMCID: PMC10052634 DOI: 10.3390/ijms24065479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/01/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Chronic stress causes several pain conditions including fibromyalgia. Its pathophysiological mechanisms are unknown, and the therapy is unresolved. Since the involvement of interleukin-1 (IL-1) has been described in stress and inflammatory pain but no data are available regarding stress-induced pain, we studied its role in a chronic restraint stress (CRS) mouse model. Female and male C57Bl/6J wild-type (WT) and IL-1αβ-deficient (knock-out: IL-1 KO) mice were exposed to 6 h of immobilization/day for 4 weeks. Mechanonociception, cold tolerance, behavioral alterations, relative thymus/adrenal gland weights, microglia ionized calcium-binding adaptor molecule 1 (IBA1) and astrocyte glial fibrillary acidic protein (GFAP) integrated density, number and morphological transformation in pain-related brain regions were determined. CRS induced 15–20% mechanical hyperalgesia after 2 weeks in WT mice in both sexes, which was significantly reduced in female but not in male IL-1 KOs. Increased IBA1+ integrated density in the central nucleus of amygdala, primary somatosensory cortex hind limb representation part, hippocampus cornu ammonis area 3 (CA3) and periaqueductal gray matter (PAG) was present, accompanied by a cell number increase in IBA1+ microglia in stressed female WTs but not in IL-1 KOs. CRS induced morphological changes of GFAP+ astrocytes in WT but not in KO mice. Stress evoked cold hypersensitivity in the stressed animals. Anxiety and depression-like behaviors, thymus and adrenal gland weight changes were detectable in all groups after 2 but not 4 weeks of CRS due to adaptation. Thus, IL-1 mediates chronic stress-induced hyperalgesia in female mice, without other major behavioral alterations, suggesting the analgesic potentials of IL-1 in blocking drugs in stress-related pain syndromes.
Collapse
Affiliation(s)
- Barbara Fülöp
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Ágnes Hunyady
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- GSK Vaccines Institute for Global Health, I-53100 Siena, Italy
| | - Noémi Bencze
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Nikolett Szentes
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Ádám Dénes
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, H-1083 Budapest, Hungary
| | - Nikolett Lénárt
- “Momentum” Laboratory of Neuroimmunology, Institute of Experimental Medicine, H-1083 Budapest, Hungary
| | - Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- Correspondence:
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre of Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- Eotvos Lorand Research Network, Chronic Pain Research Group, University of Pécs, H-7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary
| |
Collapse
|
25
|
Bando K, Tanaka Y, Winias S, Sugawara S, Mizoguchi I, Endo Y. IL-33 induces histidine decarboxylase, especially in c-kit + cells and mast cells, and roles of histamine include negative regulation of IL-33-induced eosinophilia. Inflamm Res 2023; 72:651-667. [PMID: 36723628 DOI: 10.1007/s00011-023-01699-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE AND METHODS IL-33 is present in endothelial, epithelial, and fibroblast-like cells and released upon cell injury. IL-33 reportedly induces mast-cell degranulation and is involved in various diseases, including allergic diseases. So, IL-33-related diseases seem to overlap with histamine-related diseases. In addition to the release from mast cells, histamine is newly formed by the induction of histidine decarboxylase (HDC). Some inflammatory and/or hematopoietic cytokines (IL-1, IL-3, etc.) are known to induce HDC, and the histamine produced by HDC induction is released without storage. We examined the involvement of HDC and histamine in the effects of IL-33. RESULTS A single intraperitoneal injection of IL-33 into mice induced HDC directly and/or via other cytokines (including IL-5) within a few hours in various tissues, particularly strongly in hematopoietic organs. The major cells exhibiting HDC-induction were mast cells and c-kit+ cells in the bone marrow. HDC was also induced in non-mast cells in non-hematopoietic organs. HDC, histamine, and histamine H4 receptors (H4Rs) contributed to the suppression of IL-33-induced eosinophilia. CONCLUSION IL-33 directly and indirectly (via IL-5) induces HDC in various cells, particularly potently in c-kit+ cells and mature mast cells, and the newly formed histamine contributes to the negative regulation of IL-33-induced eosinophilia via H4Rs.
Collapse
Affiliation(s)
- Kanan Bando
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan.
| | - Yukinori Tanaka
- Division of Dento-Oral Anesthesiology, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Saka Winias
- Division of Dento-Oral Anesthesiology, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Shunji Sugawara
- Division of Oral Immunology, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-Machi, Sendai, 980-8575, Japan
| | - Itaru Mizoguchi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Yasuo Endo
- Division of Oral and Maxillofacial Surgery, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-Machi, Sendai, 980-8575, Japan
| |
Collapse
|
26
|
Ashino T, Nakamura Y, Ohtaki H, Iwakura Y, Numazawa S. Interleukin-6 regulates the expression of hepatic canalicular efflux drug transporters after cecal ligation and puncture-induced sepsis: A comparison with lipopolysaccharide treatment. Toxicol Lett 2023; 374:40-47. [PMID: 36526125 DOI: 10.1016/j.toxlet.2022.12.003] [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: 10/03/2022] [Revised: 12/01/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Hepatic multidrug transporters expressed on the canalicular membrane play a role in the hepatobiliary excretion of xenobiotics and endogenous substrates. The aim of this study was to elucidate the role of pro-inflammatory cytokines in the regulation of hepatic drug transporter expression after cecal ligation and puncture (CLP), a valuable tool for studying polymicrobial sepsis, and to compare CLP with lipopolysaccharide (LPS) treatment. CLP reduced the expression of Mdr2/Abcb4, Mrp2/Abcc2, Bsep/Abcb11, Bcrp/Abcg2, and Mate1/Slc47a1 mRNAs in wild-type (WT) mouse livers in a time-dependent manner up to 48 h postoperation. LPS also reduced the expression of all transporters in WT mouse livers 24 h posttreatment; thereafter, expression levels tended to return to normal by 48 h posttreatment. IL-6-/- mice exhibited inhibited downregulation of drug transporters following CLP, although IL-1-/- and TNFα-/- mice exhibited the reduced expression of all transporters in a manner similar to that found in WT mice. Compared with CLP, LPS treatment reduced the expression of all transporters in all cytokine-deficient mouse livers, except for the expression of Mrp2/Abcc2 in IL-6-/- mice. Overall, these findings suggest that IL-6 is major factor in the downregulation of hepatic multidrug transporters following the onset of polymicrobial sepsis but not after LPS treatment.
Collapse
Affiliation(s)
- Takashi Ashino
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Yuki Nakamura
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Center for Pharmaceutical Education, Faculty of Pharmacy, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama, Kanagawa 245-0066, Japan
| | - Hirokazu Ohtaki
- Department of Functional Neurobiology, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba 278-0022, Japan
| | - Satoshi Numazawa
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| |
Collapse
|
27
|
Kaji T, Kuroishi T, Bando K, Takahashi M, Sugawara S. N-acetyl cysteine inhibits IL-1α release from murine keratinocytes induced by 2-hydroxyethyl methacrylate. J Toxicol Sci 2023; 48:557-569. [PMID: 37778984 DOI: 10.2131/jts.48.557] [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] [Indexed: 10/03/2023]
Abstract
The hydrophilic compound 2-hydroxyethyl methacrylate (HEMA) is a major component of dental bonding materials, and it enhances the binding of resin-composites to biomolecules. However, HEMA is a well-known contact sensitizer. We reported previously that intradermal injection of HEMA induces the production of IL-1 locally in the skin. Keratinocytes are the first barrier against chemical insults and constitutively express IL-1α. In this study, we analyzed whether HEMA induces the production of inflammatory cytokines from murine keratinocyte cell line Pam212 cells. We demonstrated that HEMA induced the release of 17-kDa mature IL-1α and caused cytotoxicity. The activity of calpain, an IL-1α processing enzyme, was significantly higher in HEMA-treated cells. The thiol-containing antioxidant N-acetyl cysteine (NAC) inhibited HEMA-induced IL-1α release but not cytotoxicity. NAC inhibited intracellular calpain activity and reactive oxygen species (ROS) production induced by HEMA. NAC post-treatment also inhibited IL-1α release and intracellular ROS production induced by HEMA. Furthermore, HEMA-induced in vivo inflammation also inhibited by NAC. NAC inhibited polymerization of HEMA through adduct formation via sulfide bonds between the thiol group of NAC and the reactive double bond of HEMA. HEMA-induced IL-1α release and cytotoxicity were also inhibited if HEMA and NAC were pre-incubated before adding to the cells. These results suggested that NAC inhibited IL-1α release through decreases in intracellular ROS and the adduct formation with HEMA. We concluded that HEMA induces IL-1α release from skin keratinocytes, and NAC may be a promising candidate as a therapeutic agent against inflammation induced by HEMA.
Collapse
Affiliation(s)
- Takahiro Kaji
- Division of Oral Immunology, Tohoku University Graduate School of Dentistry
| | - Toshinobu Kuroishi
- Division of Oral Immunology, Tohoku University Graduate School of Dentistry
| | - Kanan Bando
- Division of Oral Immunology, Tohoku University Graduate School of Dentistry
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry
| | - Masatoshi Takahashi
- Division of Dental Biomaterials, Tohoku University Graduate School of Dentistry
| | - Shunji Sugawara
- Division of Oral Immunology, Tohoku University Graduate School of Dentistry
| |
Collapse
|
28
|
Virgilio T, Bordini J, Cascione L, Sartori G, Latino I, Molina Romero D, Leoni C, Akhmedov M, Rinaldi A, Arribas AJ, Morone D, Seyed Jafari SM, Bersudsky M, Ottolenghi A, Kwee I, Chiaravalli AM, Sessa F, Hunger RE, Bruno A, Mortara L, Voronov E, Monticelli S, Apte RN, Bertoni F, Gonzalez SF. Subcapsular Sinus Macrophages Promote Melanoma Metastasis to the Sentinel Lymph Nodes via an IL1α-STAT3 Axis. Cancer Immunol Res 2022; 10:1525-1541. [PMID: 36206577 PMCID: PMC9716256 DOI: 10.1158/2326-6066.cir-22-0225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/18/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
During melanoma metastasis, tumor cells originating in the skin migrate via lymphatic vessels to the sentinel lymph node (sLN). This process facilitates tumor cell spread across the body. Here, we characterized the innate inflammatory response to melanoma in the metastatic microenvironment of the sLN. We found that macrophages located in the subcapsular sinus (SS) produced protumoral IL1α after recognition of tumoral antigens. Moreover, we confirmed that the elimination of LN macrophages or the administration of an IL1α-specific blocking antibody reduced metastatic spread. To understand the mechanism of action of IL1α in the context of the sLN microenvironment, we applied single-cell RNA sequencing to microdissected metastases obtained from animals treated with the IL1α-specific blocking antibody. Among the different pathways affected, we identified STAT3 as one of the main targets of IL1α signaling in metastatic tumor cells. Moreover, we found that the antitumoral effect of the anti-IL1α was not mediated by lymphocytes because Il1r1 knockout mice did not show significant differences in metastasis growth. Finally, we found a synergistic antimetastatic effect of the combination of IL1α blockade and STAT3 inhibition with stattic, highlighting a new immunotherapy approach to preventing melanoma metastasis.
Collapse
Affiliation(s)
- Tommaso Virgilio
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Joy Bordini
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,GenomSys SA, Lugano, Switzerland
| | - Luciano Cascione
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Giulio Sartori
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Irene Latino
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Daniel Molina Romero
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Graduate School Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cristina Leoni
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Murodzhon Akhmedov
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,BigOmics Analytics, Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Alberto J. Arribas
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Diego Morone
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - S. Morteza Seyed Jafari
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marina Bersudsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Aner Ottolenghi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ivo Kwee
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,BigOmics Analytics, Lugano, Switzerland
| | - Anna Maria Chiaravalli
- Unit of Pathology, ASST dei Sette Laghi, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Fausto Sessa
- Unit of Pathology, ASST dei Sette Laghi, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Robert E. Hunger
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy.,Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Silvia Monticelli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Ron N. Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Francesco Bertoni
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland.,Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Santiago F. Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Corresponding Author: Santiago F. Gonzalez, Institute for Research in Biomedicine, via Francesco Chiesa 5. CH-6500 Bellinzona. Switzerland. Phone: +41 58 666 7226; E-mail:
| |
Collapse
|
29
|
Malireddi RS, Bynigeri RR, Kancharana B, Sharma BR, Burton AR, Pelletier S, Kanneganti TD. Determining distinct roles of IL-1α through generation of an IL-1α knockout mouse with no defect in IL-1β expression. Front Immunol 2022; 13:1068230. [PMID: 36505497 PMCID: PMC9729281 DOI: 10.3389/fimmu.2022.1068230] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
Interleukin 1α (IL-1α) and IL-1β are the founding members of the IL-1 cytokine family, and these innate immune inflammatory mediators are critically important in health and disease. Early studies on these molecules suggested that their expression was interdependent, with an initial genetic model of IL-1α depletion, the IL-1α KO mouse (Il1a-KOline1), showing reduced IL-1β expression. However, studies using this line in models of infection and inflammation resulted in contrasting observations. To overcome the limitations of this genetic model, we have generated and characterized a new line of IL-1α KO mice (Il1a-KOline2) using CRISPR-Cas9 technology. In contrast to cells from Il1a-KOline1, where IL-1β expression was drastically reduced, bone marrow-derived macrophages (BMDMs) from Il1a-KOline2 mice showed normal induction and activation of IL-1β. Additionally, Il1a-KOline2 BMDMs showed normal inflammasome activation and IL-1β expression in response to multiple innate immune triggers, including both pathogen-associated molecular patterns and pathogens. Moreover, using Il1a-KOline2 cells, we confirmed that IL-1α, independent of IL-1β, is critical for the expression of the neutrophil chemoattractant KC/CXCL1. Overall, we report the generation of a new line of IL-1α KO mice and confirm functions for IL-1α independent of IL-1β. Future studies on the unique functions of IL-1α and IL-1β using these mice will be critical to identify new roles for these molecules in health and disease and develop therapeutic strategies.
Collapse
|
30
|
Rahman MFU, Yang Y, Le BT, Dutta A, Posyniak J, Faughnan P, Sayem MA, Aguilera NS, Mohi G. Interleukin-1 contributes to clonal expansion and progression of bone marrow fibrosis in JAK2V617F-induced myeloproliferative neoplasm. Nat Commun 2022; 13:5347. [PMID: 36100596 PMCID: PMC9470702 DOI: 10.1038/s41467-022-32928-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/24/2022] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation is frequently associated with myeloproliferative neoplasms (MPN), but the role of inflammation in the pathogenesis of MPN remains unclear. Expression of the proinflammatory cytokine interleukin-1 (IL-1) is elevated in patients with MPN as well as in Jak2V617F knock-in mice. Here, we show that genetic deletion of IL-1 receptor 1 (IL-1R1) normalizes peripheral blood counts, reduces splenomegaly and ameliorates bone marrow fibrosis in homozygous Jak2V617F mouse model of myelofibrosis. Deletion of IL-1R1 also significantly reduces Jak2V617F mutant hematopoietic stem/progenitor cells. Exogenous administration of IL-1β enhances myeloid cell expansion and accelerates the development of bone marrow fibrosis in heterozygous Jak2V617F mice. Furthermore, treatment with anti-IL-1R1 antibodies significantly reduces leukocytosis and splenomegaly, and ameliorates bone marrow fibrosis in homozygous Jak2V617F mice. Collectively, these results suggest that IL-1 signaling plays a pathogenic role in MPN disease progression, and targeting of IL-1R1 could be a useful strategy for the treatment of myelofibrosis.
Collapse
Affiliation(s)
- Mohammed Ferdous-Ur Rahman
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Yue Yang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Bao T Le
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Avik Dutta
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Julia Posyniak
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Patrick Faughnan
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Mohammad A Sayem
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Nadine S Aguilera
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Golam Mohi
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
- University of Virginia Cancer Center, Charlottesville, VA, 22908, USA.
| |
Collapse
|
31
|
Rai S, Grockowiak E, Hansen N, Luque Paz D, Stoll CB, Hao-Shen H, Mild-Schneider G, Dirnhofer S, Farady CJ, Méndez-Ferrer S, Skoda RC. Inhibition of interleukin-1β reduces myelofibrosis and osteosclerosis in mice with JAK2-V617F driven myeloproliferative neoplasm. Nat Commun 2022; 13:5346. [PMID: 36100613 PMCID: PMC9470591 DOI: 10.1038/s41467-022-32927-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/24/2022] [Indexed: 12/17/2022] Open
Abstract
Interleukin-1β (IL-1β) is a master regulator of inflammation. Increased activity of IL-1β has been implicated in various pathological conditions including myeloproliferative neoplasms (MPNs). Here we show that IL-1β serum levels and expression of IL-1 receptors on hematopoietic progenitors and stem cells correlate with JAK2-V617F mutant allele fraction in peripheral blood of patients with MPN. We show that the source of IL-1β overproduction in a mouse model of MPN are JAK2-V617F expressing hematopoietic cells. Knockout of IL-1β in hematopoietic cells of JAK2-V617F mice reduces inflammatory cytokines, prevents damage to nestin-positive niche cells and reduces megakaryopoiesis, resulting in decrease of myelofibrosis and osteosclerosis. Inhibition of IL-1β in JAK2-V617F mutant mice by anti-IL-1β antibody also reduces myelofibrosis and osteosclerosis and shows additive effects with ruxolitinib. These results suggest that inhibition of IL-1β with anti-IL-1β antibody alone or in combination with ruxolitinib could have beneficial effects on the clinical course in patients with myelofibrosis.
Collapse
Affiliation(s)
- Shivam Rai
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Elodie Grockowiak
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Nils Hansen
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Damien Luque Paz
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Cedric B Stoll
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Hui Hao-Shen
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Gabriele Mild-Schneider
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Stefan Dirnhofer
- Department of Pathology, University Hospital Basel, 4031, Basel, Switzerland
| | | | - Simón Méndez-Ferrer
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, CB2 0AW, UK
- Department of Hematology, University of Cambridge, Cambridge, CB2 0AW, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Radek C Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland.
| |
Collapse
|
32
|
Salmon M, Hawkins RB, Dahl J, Scott E, Johnston WF, Ailawadi G. Genetic and Pharmacological Disruption of Interleukin-1α Leads to Augmented Murine Aortic Aneurysm. Ann Vasc Surg 2022; 85:358-370. [PMID: 35680012 PMCID: PMC11029039 DOI: 10.1016/j.avsg.2022.05.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/28/2022] [Accepted: 05/11/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND Interleukin-1 (IL-1) signaling has an established role as a cytokine signaling pathway important for progression of abdominal aortic aneurysms (AAAs). While the IL-1β ligand and IL-1R1 have been previously investigated, the role of the IL-1α ligand in AAAs remains unknown. In this study, we sought to examine the role of IL-1α in AAAs using genetic and pharmacologic approaches. METHODS Eight-week-old wild-type (WT) or IL-1α knock-out (KO) male and female mice (n = 10-16/group) underwent experimental AAA and were harvested 14 days following surgery to assess AAA size and characteristics. In separate studies, 8-week-old WT mice were treated with an inhibitor to IL-1α during AAA formation and harvested 14 days following surgery. Finally, WT and IL-1α KO mice were administered Anakinra, an IL-R1 inhibitor, during AAA formation to determine the effect of inhibiting IL-1R1 when IL-1α is knocked out. RESULTS Male and female IL-1α KO mice had larger AAAs compared to WT AAAs (male: 153% vs. 89.2%, P = 0.0001; female: 86.6% vs. 63.5%, P = 0.02). IL-1α KO mice had greater elastin breakage (P = 0.01), increased levels of macrophage staining (P = 0.0045), and greater pro-metallo proteinase 2 (P = 0.02). Pharmacologic inhibition of WT male mice with an IL-1α neutralizing antibody resulted in larger AAAs (133.1% vs. 77.0%, P < 0.001). Finally, treatment of IL-1α KO male mice with Anakinra decreased AAA formation compared with vehicle control AAAs (Anakinra + IL-1α KO: 47.7% vs. WT: 147.1%; P = 0.0001). CONCLUSIONS IL-1α disruption using either genetic or pharmacologic approaches worsens AAAs.
Collapse
Affiliation(s)
- Morgan Salmon
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA; Department of Cardiac Surgery, University of Michigan School of Medicine, Ann Arbor, MI; Frankel Cardiovascular Center, University of Michigan School of Medicine, Ann Arbor, MI; Department of Surgery, Oschner Medical Center, New Orleans, LA.
| | - Robert B Hawkins
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA; Department of Cardiac Surgery, University of Michigan School of Medicine, Ann Arbor, MI; Frankel Cardiovascular Center, University of Michigan School of Medicine, Ann Arbor, MI
| | - Jolian Dahl
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA
| | - Erik Scott
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA
| | | | - Gorav Ailawadi
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA; Department of Cardiac Surgery, University of Michigan School of Medicine, Ann Arbor, MI; Frankel Cardiovascular Center, University of Michigan School of Medicine, Ann Arbor, MI
| |
Collapse
|
33
|
Tumor Temperature: Friend or Foe of Virus-Based Cancer Immunotherapy. Biomedicines 2022; 10:biomedicines10082024. [PMID: 36009571 PMCID: PMC9405776 DOI: 10.3390/biomedicines10082024] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
The temperature of a solid tumor is often dissimilar to baseline body temperature and, compared to healthy tissues, may be elevated, reduced, or a mix of both. The temperature of a tumor is dependent on metabolic activity and vascularization and can change due to tumor progression, treatment, or cancer type. Despite the need to function optimally within temperature-variable tumors, oncolytic viruses (OVs) are primarily tested at 37 °C in vitro. Furthermore, animal species utilized to test oncolytic viruses, such as mice, dogs, cats, and non-human primates, poorly recapitulate the temperature profile of humans. In this review, we discuss the importance of temperature as a variable for OV immunotherapy of solid tumors. Accumulating evidence supports that the temperature sensitivity of OVs lies on a spectrum, with some OVs likely hindered but others enhanced by elevated temperatures. We suggest that in vitro temperature sensitivity screening be performed for all OVs destined for the clinic to identify potential hinderances or benefits with regard to elevated temperature. Furthermore, we provide recommendations for the clinical use of temperature and OVs.
Collapse
|
34
|
Ratitong B, Marshall ME, Dragan MA, Anunciado CM, Abbondante S, Pearlman E. Differential Roles for IL-1α and IL-1β in Pseudomonas aeruginosa Corneal Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:548-558. [PMID: 35851538 PMCID: PMC9922050 DOI: 10.4049/jimmunol.2200110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/19/2022] [Indexed: 01/04/2023]
Abstract
Pseudomonas aeruginosa is an important cause of dermal, pulmonary, and ocular disease. Our studies have focused on P. aeruginosa infections of the cornea (keratitis) as a major cause of blinding microbial infections. The infection leads to an influx of innate immune cells, with neutrophils making up to 90% of recruited cells during early stages. We previously reported that the proinflammatory cytokines IL-1α and IL-1β were elevated during infection. Compared with wild-type (WT), infected Il1b-/- mice developed more severe corneal disease that is associated with impaired bacterial killing as a result of defective neutrophil recruitment. We also reported that neutrophils are an important source of IL-1α and IL-1β, which peaked at 24 h postinfection. To examine the role of IL-1α compared with IL-1β in P. aeruginosa keratitis, we inoculated corneas of C57BL/6 (WT), Il1a-/-, Il1b-/-, and Il1a-/-Il1b-/- (double-knockout) mice with 5 × 104 ExoS-expressing P. aeruginosa. Il1b-/- and double-knockout mice have significantly higher bacterial burden that was consistent with delayed neutrophil and monocyte recruitment to the corneas. Surprisingly, Il1a-/- mice had the opposite phenotype with enhanced bacteria clearance compared with WT mice. Although there were no significant differences in neutrophil recruitment, Il1a-/- neutrophils displayed a more proinflammatory transcriptomic profile compared to WT with elevations in C1q expression that likely caused the phenotypic differences observed. To our knowledge, our findings identify a novel, non-redundant role for IL-1α in impairing bacterial clearance.
Collapse
Affiliation(s)
- Bridget Ratitong
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA
- Institute for Immunology, University of California, Irvine, Irvine, CA
| | - Michaela E Marshall
- Department of Ophthalmology, University of California, Irvine, Irvine, CA; and
| | - Morgan A Dragan
- Institute for Immunology, University of California, Irvine, Irvine, CA
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA
| | - Charissa M Anunciado
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA
| | - Serena Abbondante
- Department of Ophthalmology, University of California, Irvine, Irvine, CA; and
| | - Eric Pearlman
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA
- Institute for Immunology, University of California, Irvine, Irvine, CA
- Department of Ophthalmology, University of California, Irvine, Irvine, CA; and
| |
Collapse
|
35
|
Hayer S, Niederreiter B, Kalkgruber M, Wanic K, Maißner J, Smolen JS, Aletaha D, Blüml S, Redlich K. Analysis of combined deficiency of interleukin-1 and -6 versus single deficiencies in TNF-mediated arthritis and systemic bone loss. Bone Joint Res 2022; 11:484-493. [PMID: 35801532 PMCID: PMC9350695 DOI: 10.1302/2046-3758.117.bjr-2021-0481.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Aims Insufficient treatment response in rheumatoid arthritis (RA) patients requires novel treatment strategies to halt disease progression. The potential benefit of combination of cytokine-inhibitors in RA is still unclear and needs further investigation. To explore the impact of combined deficiency of two major cytokines, namely interleukin (IL)-1 and IL-6, in this study double deficient mice for IL-1αβ and IL-6 were investigated in different tumour necrosis factor (TNF)-driven inflammatory bone disorders, namely peripheral arthritis and sacroiliitis, as well as systemic bone loss. Methods Disease course, histopathological features of arthritis, and micro-CT (µCT) bone analysis of local and systemic bone loss were assessed in 15-week-old IL1-/-IL6-/-hTNFtg in comparison to IL1-/-hTNFtg, IL6-/-hTNFtg, and hTNFtg mice. µCT bone analysis of single deficient and wild-type mice was also performed. Results Combined deficiency of IL-1/IL-6 markedly ameliorated TNF-mediated arthritis and bilateral sacroiliitis, but without additive benefits compared to single IL-1 deficiency. This finding confirms the important role of IL-1 and the marginal role of IL-6 in TNF-driven pathways of local joint damage, but questions the efficacy of potential combinatorial therapies of IL-1 and IL-6 in treatment of RA. In contrast, combined deficiency of IL-1/IL-6 led to an additive protective effect on TNF-driven systemic bone loss compared to single IL-1 and IL-6 deficiency. This finding clearly indicates a common contribution of both IL-1 and IL-6 in TNF-driven systemic bone loss, and points to a discrepancy of cytokine dependency in local and systemic TNF-driven mechanisms of inflammatory arthritis. Conclusion Combinatorial treatments in RA might provide different benefits to inflammatory local arthritis and systemic comorbidities. Cite this article: Bone Joint Res 2022;11(7):484–493.
Collapse
Affiliation(s)
- Silvia Hayer
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Birgit Niederreiter
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Marlene Kalkgruber
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Konrad Wanic
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Julia Maißner
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Stephan Blüml
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Kurt Redlich
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.,Hietzing Hospital, 2nd Department of Medicine, Vienna, Austria.,Karl Landsteiner Institute for Rheumatology and Clinical Immunology, Vienna, Austria
| |
Collapse
|
36
|
Yamagishi R, Kamachi F, Nakamura M, Yamazaki S, Kamiya T, Takasugi M, Cheng Y, Nonaka Y, Yukawa-Muto Y, Thuy LTT, Harada Y, Arai T, Loo TM, Yoshimoto S, Ando T, Nakajima M, Taguchi H, Ishikawa T, Akiba H, Miyake S, Kubo M, Iwakura Y, Fukuda S, Chen WY, Kawada N, Rudensky A, Nakae S, Hara E, Ohtani N. Gasdermin D-mediated release of IL-33 from senescent hepatic stellate cells promotes obesity-associated hepatocellular carcinoma. Sci Immunol 2022; 7:eabl7209. [PMID: 35749514 DOI: 10.1126/sciimmunol.abl7209] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Long-term senescent cells exhibit a secretome termed the senescence-associated secretory phenotype (SASP). Although the mechanisms of SASP factor induction have been intensively studied, the release mechanism and how SASP factors influence tumorigenesis in the biological context remain unclear. In this study, using a mouse model of obesity-induced hepatocellular carcinoma (HCC), we identified the release mechanism of SASP factors, which include interleukin-1β (IL-1β)- and IL-1β-dependent IL-33, from senescent hepatic stellate cells (HSCs) via gasdermin D (GSDMD) amino-terminal-mediated pore. We found that IL-33 was highly induced in senescent HSCs in an IL-1β-dependent manner in the tumor microenvironment. The release of both IL-33 and IL-1β was triggered by lipoteichoic acid (LTA), a cell wall component of gut microbiota that was transferred and accumulated in the liver tissue of high-fat diet-fed mice, and the release of these factors was mediated through cell membrane pores formed by the GSDMD amino terminus, which was cleaved by LTA-induced caspase-11. We demonstrated that IL-33 release from HSCs promoted HCC development via the activation of ST2-positive Treg cells in the liver tumor microenvironment. The accumulation of GSDMD amino terminus was also detected in HSCs from human NASH-associated HCC patients, suggesting that similar mechanism could be involved in a certain type of human HCC. These results uncover a release mechanism for SASP factors from sensitized senescent HSCs in the tumor microenvironment, thereby facilitating obesity-associated HCC progression. Furthermore, our findings highlight the therapeutic potential of inhibitors of GSDMD-mediated pore formation for HCC treatment.
Collapse
Affiliation(s)
- Ryota Yamagishi
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University)
| | - Fumitaka Kamachi
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University).,Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | - Masaru Nakamura
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | - Shota Yamazaki
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | - Tomonori Kamiya
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University)
| | - Masaki Takasugi
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University)
| | - Yi Cheng
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University)
| | - Yoshiki Nonaka
- Department of Pathophysiology, Osaka City University, Graduate School of Medicine, Osaka, Japan
| | - Yoshimi Yukawa-Muto
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University).,Department of Hepatology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University)
| | - Le Thi Thanh Thuy
- Department of Hepatology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University)
| | - Yohsuke Harada
- Laboratory of Pharmaceutical Immunology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Tatsuya Arai
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | - Tze Mun Loo
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan.,Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Shin Yoshimoto
- Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Tatsuya Ando
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | - Masahiro Nakajima
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | - Hayao Taguchi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | - Takamasa Ishikawa
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Hisaya Akiba
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Masato Kubo
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda, Chiba, Japan.,Laboratory for Cytokine Regulation, Research Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Kanagawa, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan.,Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan.,Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Wei-Yu Chen
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Norifumi Kawada
- Department of Hepatology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University)
| | - Alexander Rudensky
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susumu Nakae
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima City, Hiroshima, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| | - Eiji Hara
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan.,Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Naoko Ohtani
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan (formerly, Osaka City University).,Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
37
|
Sterling JK, Baumann B, Foshe S, Voigt A, Guttha S, Alnemri A, McCright SJ, Li M, Zauhar RJ, Montezuma SR, Kapphahn RJ, Chavali VRM, Hill DA, Ferrington DA, Stambolian D, Mullins RF, Merrick D, Dunaief JL. Inflammatory adipose activates a nutritional immunity pathway leading to retinal dysfunction. Cell Rep 2022; 39:110942. [PMID: 35705048 PMCID: PMC9248858 DOI: 10.1016/j.celrep.2022.110942] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 01/24/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022] Open
Abstract
Age-related macular degeneration (AMD), the leading cause of irreversible blindness among Americans over 50, is characterized by dysfunction and death of retinal pigment epithelial (RPE) cells. The RPE accumulates iron in AMD, and iron overload triggers RPE cell death in vitro and in vivo. However, the mechanism of RPE iron accumulation in AMD is unknown. We show that high-fat-diet-induced obesity, a risk factor for AMD, drives systemic and local inflammatory circuits upregulating interleukin-1β (IL-1β). IL-1β upregulates RPE iron importers and downregulates iron exporters, causing iron accumulation, oxidative stress, and dysfunction. We term this maladaptive, chronic activation of a nutritional immunity pathway the cellular iron sequestration response (CISR). RNA sequencing (RNA-seq) analysis of choroid and retina from human donors revealed that hallmarks of this pathway are present in AMD microglia and macrophages. Together, these data suggest that inflamed adipose tissue, through the CISR, can lead to RPE pathology in AMD.
Collapse
Affiliation(s)
- Jacob K Sterling
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Bailey Baumann
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sierra Foshe
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Andrew Voigt
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Samyuktha Guttha
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ahab Alnemri
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sam J McCright
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Randy J Zauhar
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, PA 19104, USA
| | - Sandra R Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rebecca J Kapphahn
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Venkata R M Chavali
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David A Hill
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Deborah A Ferrington
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dwight Stambolian
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Robert F Mullins
- Institute for Vision Research, The University of Iowa, Iowa City, IA 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - David Merrick
- Department of Medicine, Division of Endocrinology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joshua L Dunaief
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
38
|
Adachi A, Honda T, Egawa G, Kanameishi S, Takimoto R, Miyake T, Hossain MR, Komine M, Ohtsuki M, Gunzer M, Ikuta K, Kabashima K. Estradiol suppresses psoriatic inflammation in mice by regulating neutrophil and macrophage functions. J Allergy Clin Immunol 2022; 150:909-919.e8. [PMID: 35589416 DOI: 10.1016/j.jaci.2022.03.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/21/2022] [Accepted: 03/18/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Psoriasis is a common inflammatory skin disease resulting from dysregulation of the IL-23/TH17 immune axis. The prevalence and severity of psoriasis is higher in men than in women, although the underlying reasons for this are unclear. OBJECTIVE We studied whether estradiol, a female hormone, plays protective roles in imiquimod-induced psoriatic inflammation in mice by regulating neutrophil and macrophage functions. METHODS Wild-type mice and conditional knockout mice were ovariectomized, supplemented with placebo or estradiol pellets, and an imiquimod-containing cream applied. RESULTS Mice without endogenous ovarian hormones exhibited exacerbated psoriatic inflammation including increased production of IL-17A and IL-1β, which was reversed by exogenously added estradiol. The suppressive effect of estradiol on the production of IL-1β and IL-17A was abolished in mice lacking estrogen receptors in neutrophils and macrophages (Esr1f/fEsr2f/fLysM-Cre+ mice). IL-1β, which is required for production of IL-17A in the psoriasis model, was mainly produced by neutrophils and inflammatory macrophages. Estradiol suppressed IL-1β production from neutrophils and macrophages in mice both in vivo and in vitro and from human neutrophils in vitro. CONCLUSION Our results suggest a novel mechanism for sex-dependent differences in psoriasis clinical phenotypes that may shed new light on the pathology of psoriasis.
Collapse
Affiliation(s)
- Akimasa Adachi
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Dermatology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Tetsuya Honda
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Gyohei Egawa
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuto Kanameishi
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Riko Takimoto
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshiya Miyake
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Md Razib Hossain
- Department of Dermatology, Jichi Medical University Graduate School of Medicine, Shimotsuke, Japan
| | - Mayumi Komine
- Department of Dermatology, Jichi Medical University Graduate School of Medicine, Shimotsuke, Japan
| | - Mamitaro Ohtsuki
- Department of Dermatology, Jichi Medical University Graduate School of Medicine, Shimotsuke, Japan
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany; Leibniz-Institut für Analytische Wissenschaften ISAS-e.V, Dortmund, Germany
| | - Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Singapore Immunology Network (SIgN) and Skin Research Institute of Singapore (SRIS), Technology and Research (A∗STAR), Singapore.
| |
Collapse
|
39
|
Schädlich IS, Vienhues JH, Jander A, Piepke M, Magnus T, Lambertsen KL, Clausen BH, Gelderblom M. Interleukin-1 Mediates Ischemic Brain Injury via Induction of IL-17A in γδ T Cells and CXCL1 in Astrocytes. Neuromolecular Med 2022; 24:437-451. [PMID: 35384588 PMCID: PMC9684245 DOI: 10.1007/s12017-022-08709-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/04/2022] [Indexed: 11/29/2022]
Abstract
As a prototypical proinflammatory cytokine, interleukin-1 (IL-1) exacerbates the early post-stroke inflammation, whereas its neutralization is protective. To further investigate the underlying cell-type-specific IL-1 effects, we subjected IL-1 (α/β) knockout (Il1−/−) and wildtype (WT) littermate mice to permanent middle cerebral artery occlusion (pMCAO) and assessed immune cell infiltration and cytokine production in the ischemic hemisphere by flow cytometry 24 h and 72 h after stroke. Il1−/− mice showed smaller infarcts and reduced neutrophil infiltration into the ischemic brain. We identified γδ T cells and astrocytes as target cells of IL-1 signaling-mediated neutrophil recruitment. First, IL-1-induced IL-17A production in γδ T cells in vivo, and IL-17A enhanced the expression of the main neutrophil attracting chemokine CXCL1 by astrocytes in the presence of tumor necrosis factor (TNF) in vitro. Second, IL-1 itself was a potent activator of astrocytic CXCL1 production in vitro. By employing a novel FACS sorting strategy for the acute isolation of astrocytes from ischemic brains, we confirmed that IL-1 is pivotal for Cxcl1 upregulation in astrocytes in vivo. Our results underscore the pleiotropic effects of IL-1 on immune and non-immune cells within the CNS to mount and amplify the post-stroke inflammatory response.
Collapse
Affiliation(s)
- Ines Sophie Schädlich
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany.
| | - Jonas Heinrich Vienhues
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Alina Jander
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany
| | - Marius Piepke
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Deparment of Neurology, Odense University Hospital, Odense, Denmark
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg-Eppendorf, Germany
| |
Collapse
|
40
|
Eislmayr K, Bestehorn A, Morelli L, Borroni M, Walle LV, Lamkanfi M, Kovarik P. Nonredundancy of IL-1α and IL-1β is defined by distinct regulation of tissues orchestrating resistance versus tolerance to infection. SCIENCE ADVANCES 2022; 8:eabj7293. [PMID: 35235356 PMCID: PMC8890706 DOI: 10.1126/sciadv.abj7293] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 01/05/2022] [Indexed: 06/08/2023]
Abstract
Interleukin-1α (IL-1α) and IL-1β are inflammatory cytokines with important roles in health and disease. They trigger the same receptor and elicit comparable cellular responses but, for poorly understood reasons, are not redundant in vivo. Here, we decoupled IL-1α and IL-1β functions that drive protective responses against invasive infection with group A Streptococcus. IL-1β was essential for pathogen clearance, hence resistance to infection, by inducing granulocyte colony-stimulating factor at the infection site and establishing emergency granulopoiesis. In contrast, IL-1α governed reprogramming of liver metabolic pathways associated with tolerance to infection. The IL-1α-dominated hepatic regulation corresponded to high IL-1α levels in the liver during infection. Conversely, IL-1β was critical for the regulation of the spleen transcriptome, which correlated with ample IL-1β expression in this tissue. The results identify distinct and organ-specific roles of IL-1α versus IL-1β and implicate spatial restriction of their expression and bioavailability during infection as the underlying mechanism.
Collapse
Affiliation(s)
- Kevin Eislmayr
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Annika Bestehorn
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Luisa Morelli
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Martina Borroni
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| | - Lieselotte Vande Walle
- Laboratory of Medical Immunology, Department of Internal Medicine and Pediatrics, Ghent University, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Mohamed Lamkanfi
- Laboratory of Medical Immunology, Department of Internal Medicine and Pediatrics, Ghent University, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Pavel Kovarik
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, A-1030 Vienna, Austria
| |
Collapse
|
41
|
Tseng HW, Kulina I, Girard D, Gueguen J, Vaquette C, Salga M, Fleming W, Jose B, Millard SM, Pettit AR, Schroder K, Thomas G, Wheeler L, Genêt F, Banzet S, Alexander KA, Lévesque JP. Interleukin-1 Is Overexpressed in Injured Muscles Following Spinal Cord Injury and Promotes Neurogenic Heterotopic Ossification. J Bone Miner Res 2022; 37:531-546. [PMID: 34841579 DOI: 10.1002/jbmr.4482] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
Neurogenic heterotopic ossifications (NHOs) form in periarticular muscles after severe spinal cord (SCI) and traumatic brain injuries. The pathogenesis of NHO is poorly understood with no effective preventive treatment. The only curative treatment remains surgical resection of pathological NHOs. In a mouse model of SCI-induced NHO that involves a transection of the spinal cord combined with a muscle injury, a differential gene expression analysis revealed that genes involved in inflammation such as interleukin-1β (IL-1β) were overexpressed in muscles developing NHO. Using mice knocked-out for the gene encoding IL-1 receptor (IL1R1) and neutralizing antibodies for IL-1α and IL-1β, we show that IL-1 signaling contributes to NHO development after SCI in mice. Interestingly, other proteins involved in inflammation that were also overexpressed in muscles developing NHO, such as colony-stimulating factor-1, tumor necrosis factor, or C-C chemokine ligand-2, did not promote NHO development. Finally, using NHO biopsies from SCI and TBI patients, we show that IL-1β is expressed by CD68+ macrophages. IL-1α and IL-1β produced by activated human monocytes promote calcium mineralization and RUNX2 expression in fibro-adipogenic progenitors isolated from muscles surrounding NHOs. Altogether, these data suggest that interleukin-1 promotes NHO development in both humans and mice. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Hsu-Wen Tseng
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| | - Irina Kulina
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| | - Dorothée Girard
- Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France.,INSERM UMR-MD 1197, Université de Paris-Saclay, Gif-sur-Yvette, France
| | - Jules Gueguen
- Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France.,INSERM UMR-MD 1197, Université de Paris-Saclay, Gif-sur-Yvette, France
| | - Cedryck Vaquette
- School of Dentistry, The University of Queensland, Herston, Australia.,Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Australia
| | - Marjorie Salga
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia.,Unité Péri Opératoire du Handicap (UPOH), PMR Department, Versailles Saint-Quentin-en-Yvelines University (UVSQ); UFR Simone Veil - Santé, END: ICAP, INSERM U1179, Hôpital Raymond-Poincaré, Assistance Publique - Hôpitaux de Paris (AP-HP), Garches, France.,Université de Versailles Saint-Quentin-en-Yvelines (UVSQ); UFR Simone Veil - Santé, END: ICAP, INSERM U1179, Montigny-le-Bretonneux, France
| | - Whitney Fleming
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| | - Beulah Jose
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| | - Susan M Millard
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| | - Allison R Pettit
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience, University of Queensland, Saint Lucia, Australia
| | - Gethin Thomas
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Australia
| | - Lawrie Wheeler
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Australia
| | - François Genêt
- Unité Péri Opératoire du Handicap (UPOH), PMR Department, Versailles Saint-Quentin-en-Yvelines University (UVSQ); UFR Simone Veil - Santé, END: ICAP, INSERM U1179, Hôpital Raymond-Poincaré, Assistance Publique - Hôpitaux de Paris (AP-HP), Garches, France.,Université de Versailles Saint-Quentin-en-Yvelines (UVSQ); UFR Simone Veil - Santé, END: ICAP, INSERM U1179, Montigny-le-Bretonneux, France
| | - Sébastien Banzet
- Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France.,INSERM UMR-MD 1197, Université de Paris-Saclay, Gif-sur-Yvette, France
| | - Kylie A Alexander
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| | - Jean-Pierre Lévesque
- Mater Research Institute - The University of Queensland, Woolloongabba, Australia
| |
Collapse
|
42
|
Maternal P2X7 receptor inhibition prevents autism-like phenotype in male mouse offspring through the NLRP3-IL-1β pathway. Brain Behav Immun 2022; 101:318-332. [PMID: 35065198 DOI: 10.1016/j.bbi.2022.01.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/29/2021] [Accepted: 01/16/2022] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition caused by interactions of environmental and genetic factors. Recently we showed that activation of the purinergic P2X7 receptors is necessary and sufficient to convert maternal immune activation (MIA) to ASD-like features in male offspring mice. Our aim was to further substantiate these findings and identify downstream signaling pathways coupled to P2X7 upon MIA. Maternal treatment with the NLRP3 antagonist MCC950 and a neutralising IL-1β antibody during pregnancy counteracted the development of autistic characteristics in offspring mice. We also explored time-dependent changes of a widespread cytokine and chemokine profile in maternal blood and fetal brain samples of poly(I:C)/saline-treated dams. MIA-induced increases in plasma IL-1β, RANTES, MCP-1, and fetal brain IL-1β, IL-2, IL-6, MCP-1 concentrations are regulated by the P2X7/NLRP3 pathway. Offspring treatment with the selective P2X7 receptor antagonist JNJ47965567 was effective in the prevention of autism-like behavior in mice using a repeated dosing protocol. Our results highlight that in addition to P2X7, NLRP3, as well as inflammatory cytokines, may also be potential biomarkers and therapeutic targets of social deficits and repetitive behaviors observed in autism spectrum disorder.
Collapse
|
43
|
Host Lung Environment Limits Aspergillus fumigatus Germination through an SskA-Dependent Signaling Response. mSphere 2021; 6:e0092221. [PMID: 34878292 PMCID: PMC8653827 DOI: 10.1128/msphere.00922-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aspergillus fumigatus isolates display significant heterogeneity in growth, virulence, pathology, and inflammatory potential in multiple murine models of invasive aspergillosis. Previous studies have linked the initial germination of a fungal isolate in the airways to the inflammatory and pathological potential, but the mechanism(s) regulating A. fumigatus germination in the airways is unresolved. To explore the genetic basis for divergent germination phenotypes, we utilized a serial passaging strategy in which we cultured a slow germinating strain (AF293) in a murine-lung-based medium for multiple generations. Through this serial passaging approach, a strain emerged with an increased germination rate that induces more inflammation than the parental strain (herein named LH-EVOL for lung homogenate evolved). We identified a potential loss-of-function allele of Afu5g08390 (sskA) in the LH-EVOL strain. The LH-EVOL strain had a decreased ability to induce the SakA-dependent stress pathway, similar to AF293 ΔsskA and CEA10. In support of the whole-genome variant analyses, sskA, sakA, or mpkC loss-of-function strains in the AF293 parental strain increased germination both in vitro and in vivo. Since the airway surface liquid of the lungs contains low glucose levels, the relationship of low glucose concentration on germination of these mutant AF293 strains was examined; interestingly, in low glucose conditions, the sakA pathway mutants exhibited an enhanced germination rate. In conclusion, A. fumigatus germination in the airways is regulated by SskA through the SakA mitogen-activated protein kinase (MAPK) pathway and drives enhanced disease initiation and inflammation in the lungs. IMPORTANCEAspergillus fumigatus is an important human fungal pathogen particularly in immunocompromised individuals. Initiation of growth by A. fumigatus in the lung is important for its pathogenicity in murine models. However, our understanding of what regulates fungal germination in the lung environment is lacking. Through a serial passage experiment using lung-based medium, we identified a new strain of A. fumigatus that has increased germination potential and inflammation in the lungs. Using this serially passaged strain, we found it had a decreased ability to mediate signaling through the osmotic stress response pathway. This finding was confirmed using genetic null mutants demonstrating that the osmotic stress response pathway is critical for regulating growth in the murine lungs. Our results contribute to the understanding of A. fumigatus adaptation and growth in the host lung environment.
Collapse
|
44
|
Dosch AR, Singh S, Nagathihalli NS, Datta J, Merchant NB. Interleukin-1 signaling in solid organ malignancies. Biochim Biophys Acta Rev Cancer 2021; 1877:188670. [PMID: 34923027 DOI: 10.1016/j.bbcan.2021.188670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/19/2021] [Accepted: 12/10/2021] [Indexed: 12/20/2022]
Abstract
As inflammation plays a critical role in the development and progression of cancer, therapeutic targeting of cytokine pathways involved in both tumorigenesis and dictating response to clinical treatments are of significant interest. Recent evidence has highlighted the importance of the pro-inflammatory cytokine interleukin-1 (IL-1) as a key mediator of tumor growth, metastatic disease spread, immunosuppression, and drug resistance in cancer. IL-1 promotes tumorigenesis through diverse mechanisms, including the activation of oncogenic signaling pathways directly in tumor cells and via orchestrating crosstalk between the cellular constituents of the tumor microenvironment (TME), thereby driving cancer growth. This review will provide an overview of IL-1 signaling and physiology and summarize the disparate mechanisms involving IL-1 in tumorigenesis and cancer progression. Additionally, clinical studies targeting IL-1 signaling in the management of solid organ tumors will be summarized herein.
Collapse
Affiliation(s)
- Austin R Dosch
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Samara Singh
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Nagaraj S Nagathihalli
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Jashodeep Datta
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Nipun B Merchant
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America.
| |
Collapse
|
45
|
Ren WJ, Illes P. Involvement of P2X7 receptors in chronic pain disorders. Purinergic Signal 2021; 18:83-92. [PMID: 34799827 PMCID: PMC8850523 DOI: 10.1007/s11302-021-09796-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic pain is caused by cellular damage with an obligatory inflammatory component. In response to noxious stimuli, high levels of ATP leave according to their concentration gradient, the intracellular space through discontinuities generated in the plasma membrane or diffusion through pannexin-1 hemichannels, and activate P2X7Rs localized at peripheral and central immune cells. Because of the involvement of P2X7Rs in immune functions and especially the initiation of macrophage/microglial and astrocytic secretion of cytokines, chemokines, prostaglandins, proteases, reactive oxygen, and nitrogen species as well as the excitotoxic glutamate/ATP, this receptor type has a key role in chronic pain processes. Microglia are equipped with a battery of pattern recognition receptors that detect pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS) from bacterial infections or danger associated molecular patterns (DAMPs) such as ATP. The co-stimulation of these receptors leads to the activation of the NLRP3 inflammasome and interleukin-1β (IL-1β) release. In the present review, we invite you to a journey through inflammatory and neuropathic pain, primary headache, and regulation of morphine analgesic tolerance, in the pathophysiology of which P2X7Rs are centrally involved. P2X7R bearing microglia and astrocyte-like cells playing eminent roles in chronic pain will be also discussed.
Collapse
Affiliation(s)
- Wen-Jing Ren
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Peter Illes
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04109, Leipzig, Germany.
| |
Collapse
|
46
|
Hausmann A, Felmy B, Kunz L, Kroon S, Berthold DL, Ganz G, Sandu I, Nakamura T, Zangger NS, Zhang Y, Dolowschiak T, Fattinger SA, Furter M, Müller-Hauser AA, Barthel M, Vlantis K, Wachsmuth L, Kisielow J, Tortola L, Heide D, Heikenwälder M, Oxenius A, Kopf M, Schroeder T, Pasparakis M, Sellin ME, Hardt WD. Intercrypt sentinel macrophages tune antibacterial NF-κB responses in gut epithelial cells via TNF. J Exp Med 2021; 218:e20210862. [PMID: 34529751 PMCID: PMC8480669 DOI: 10.1084/jem.20210862] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/21/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal epithelial cell (IEC) NF-κB signaling regulates the balance between mucosal homeostasis and inflammation. It is not fully understood which signals tune this balance and how bacterial exposure elicits the process. Pure LPS induces epithelial NF-κB activation in vivo. However, we found that in mice, IECs do not respond directly to LPS. Instead, tissue-resident lamina propria intercrypt macrophages sense LPS via TLR4 and rapidly secrete TNF to elicit epithelial NF-κB signaling in their immediate neighborhood. This response pattern is relevant also during oral enteropathogen infection. The macrophage-TNF-IEC axis avoids responses to luminal microbiota LPS but enables crypt- or tissue-scale epithelial NF-κB responses in proportion to the microbial threat. Thereby, intercrypt macrophages fulfill important sentinel functions as first responders to Gram-negative microbes breaching the epithelial barrier. The tunability of this crypt response allows the induction of defense mechanisms at an appropriate scale according to the localization and intensity of microbial triggers.
Collapse
Affiliation(s)
- Annika Hausmann
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Boas Felmy
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Leo Kunz
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Sanne Kroon
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Dorothée Lisa Berthold
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Giverny Ganz
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Ioana Sandu
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Toshihiro Nakamura
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Nathan Sébastien Zangger
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Yang Zhang
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Tamas Dolowschiak
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Stefan Alexander Fattinger
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Markus Furter
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Anna Angelika Müller-Hauser
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Katerina Vlantis
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Laurens Wachsmuth
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Jan Kisielow
- Institute of Molecular Health Sciences, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Luigi Tortola
- Institute of Molecular Health Sciences, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Danijela Heide
- Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Annette Oxenius
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Manolis Pasparakis
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Mikael Erik Sellin
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| |
Collapse
|
47
|
Takakura Y, Hori N, Terada N, Machida M, Yamaguchi N, Takano H, Yamaguchi N. VGLL3 activates inflammatory responses by inducing interleukin-1α secretion. FASEB J 2021; 35:e21996. [PMID: 34679187 DOI: 10.1096/fj.202100679rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/05/2021] [Accepted: 10/04/2021] [Indexed: 01/10/2023]
Abstract
Vestigial-like family member 3 (VGLL3), a member of the vestigial-like family, is a cofactor of the TEA-domain-containing transcription factor (TEAD). Although elevation in VGLL3 expression is associated with inflammatory diseases, such as inflammatory sarcomas and autoimmune diseases, the molecular mechanisms underlying VGLL3-mediated inflammation remain largely unknown. In this study, we analyzed the relationship between elevated VGLL3 expression and the levels of NF-κB, a transcription factor that plays a pivotal role in inflammation. NF-κB was found to be activated in a cell line stably expressing VGLL3. Mechanistically, VGLL3 was shown to promote the expression and secretion of the potent NF-κB-activating cytokine interleukin (IL)-1α, probably through its association with TEADs. As VGLL3 is a target of transforming growth factor β (TGF-β) signaling, we analyzed IL-1α induction upon TGF-β stimulation. TGF-β stimulation was observed to induce IL-1α secretion and NF-κB activation, and VGLL3 was associated with this phenomenon. The TGF-β transcription factors Smad3 and Smad4 were shown to be necessary for inducing VGLL3 and IL-1α expression. Lastly, we found that VGLL3-dependent IL-1α secretion is involved in constitutive NF-κB activation in highly malignant breast cancer cells. Collectively, the findings suggested that VGLL3 expression and TGF-β stimulation activate the inflammatory response by inducing IL-1α secretion.
Collapse
Affiliation(s)
- Yuki Takakura
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Naoto Hori
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan.,Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Natsumi Terada
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Moeka Machida
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan.,Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Naoto Yamaguchi
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroyuki Takano
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Noritaka Yamaguchi
- Laboratory of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan.,Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
48
|
Akita K, Isoda K, Ohtomo F, Isobe S, Niida T, Sato-Okabayashi Y, Sano M, Shimada K, Iwakura Y, Minamino T. Blocking of interleukin-1 suppresses angiotensin II-induced renal injury. Clin Sci (Lond) 2021; 135:2035-2048. [PMID: 34402864 DOI: 10.1042/cs20201406] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 11/17/2022]
Abstract
Clinical hypertension (HT) is associated with renal inflammation and elevated circulating levels of proinflammatory cytokines. Interleukin (IL)-1 receptor antagonist (IL-1Ra) is one of the most important anti-inflammatory cytokines and plays a crucial role in inflammation. Inhibition of IL-1 may contribute to modulation of the Angiotensin II (Ang II)-induced HT response. The present study aimed to elucidate the effects of IL-1Ra and anti-IL-1β antibody (01BSUR) on Ang II-induced renal injury. To determine the contribution of IL-1Ra to Ang II-induced renal inflammation, male wildtype (WT) and IL-1Ra-deficient (IL-1Ra-/-) mice were infused with Ang II (1000 ng/kg/min) using subcutaneous osmotic pump for 14 days. We checked renal function, histological change, and several mRNA expressions 14 days after infusion. Fourteen days after infusion, systolic blood pressure (197 ± 5 vs 169 ± 9 mmHg, P<0.05) in IL-1Ra-/- mice significantly increased compared with WT mice. Furthermore, on day 14 of Ang II infusion, plasma IL-6 was 5.9-fold higher in IL-1Ra-/- versus WT mice (P<0.001); renal preproendothelin-1 mRNA expression was also significantly higher in IL-1Ra-/- mice (P<0.05). In addition, renal histology revealed greater damage in IL-1Ra-/- mice compared with WT mice 14 days after infusion. Finally, we administrated 01BSUR to both IL-1Ra-/- and WT mice, and 01BSUR treatment decreased Ang II-induced HT and renal damage (glomerular injury and fibrosis of the tubulointerstitial area) in both IL-1Ra-/- and WT mice compared with IgG2a treatment. Inhibition of IL-1 decreased Ang II-induced HT and renal damage in both IL-1Ra-/- and WT mice, suggesting suppression of IL-1 may provide an additional strategy to protect against renal damage in hypertensive patients.
Collapse
Affiliation(s)
- Koji Akita
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Kikuo Isoda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Cardiology, Juntendo University Nerima Hospital, Nerimaku, Tokyo, Japan
| | - Fumie Ohtomo
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Sarasa Isobe
- Division of Cardiology, Keio University, Shinanomachi, Tokyo, Japan
| | - Tomiharu Niida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yayoi Sato-Okabayashi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Motoaki Sano
- Division of Cardiology, Keio University, Shinanomachi, Tokyo, Japan
| | - Kazunori Shimada
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
49
|
Fukuda K, Okamura K, Riding RL, Fan X, Afshari K, Haddadi NS, McCauley SM, Guney MH, Luban J, Funakoshi T, Yaguchi T, Kawakami Y, Khvorova A, Fitzgerald KA, Harris JE. AIM2 regulates anti-tumor immunity and is a viable therapeutic target for melanoma. J Exp Med 2021; 218:212521. [PMID: 34325468 PMCID: PMC8329870 DOI: 10.1084/jem.20200962] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/24/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
The STING and absent in melanoma 2 (AIM2) pathways are activated by the presence of cytosolic DNA, and STING agonists enhance immunotherapeutic responses. Here, we show that dendritic cell (DC) expression of AIM2 within human melanoma correlates with poor prognosis and, in contrast to STING, AIM2 exerts an immunosuppressive effect within the melanoma microenvironment. Vaccination with AIM2-deficient DCs improves the efficacy of both adoptive T cell therapy and anti–PD-1 immunotherapy for “cold tumors,” which exhibit poor therapeutic responses. This effect did not depend on prolonged survival of vaccinated DCs, but on tumor-derived DNA that activates STING-dependent type I IFN secretion and subsequent production of CXCL10 to recruit CD8+ T cells. Additionally, loss of AIM2-dependent IL-1β and IL-18 processing enhanced the treatment response further by limiting the recruitment of regulatory T cells. Finally, AIM2 siRNA-treated mouse DCs in vivo and human DCs in vitro enhanced similar anti-tumor immune responses. Thus, targeting AIM2 in tumor-infiltrating DCs is a promising new treatment strategy for melanoma.
Collapse
Affiliation(s)
- Keitaro Fukuda
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA.,Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Ken Okamura
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Rebecca L Riding
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Xueli Fan
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Khashayar Afshari
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Nazgol-Sadat Haddadi
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| | - Sean M McCauley
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Mehmet H Guney
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA.,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA
| | - Takeru Funakoshi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA.,Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Katherine A Fitzgerald
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA
| | - John E Harris
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
50
|
Morinaga H, Mohri Y, Grachtchouk M, Asakawa K, Matsumura H, Oshima M, Takayama N, Kato T, Nishimori Y, Sorimachi Y, Takubo K, Suganami T, Iwama A, Iwakura Y, Dlugosz AA, Nishimura EK. Obesity accelerates hair thinning by stem cell-centric converging mechanisms. Nature 2021; 595:266-271. [PMID: 34163066 PMCID: PMC9600322 DOI: 10.1038/s41586-021-03624-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Obesity is a worldwide epidemic that predisposes individuals to many age-associated diseases, but its exact effects on organ dysfunction are largely unknown1. Hair follicles-mini-epithelial organs that grow hair-are miniaturized by ageing to cause hair loss through the depletion of hair follicle stem cells (HFSCs)2. Here we report that obesity-induced stress, such as that induced by a high-fat diet (HFD), targets HFSCs to accelerate hair thinning. Chronological gene expression analysis revealed that HFD feeding for four consecutive days in young mice directed activated HFSCs towards epidermal keratinization by generating excess reactive oxygen species, but did not reduce the pool of HFSCs. Integrative analysis using stem cell fate tracing, epigenetics and reverse genetics showed that further feeding with an HFD subsequently induced lipid droplets and NF-κB activation within HFSCs via autocrine and/or paracrine IL-1R signalling. These integrated factors converge on the marked inhibition of Sonic hedgehog (SHH) signal transduction in HFSCs, thereby further depleting lipid-laden HFSCs through their aberrant differentiation and inducing hair follicle miniaturization and eventual hair loss. Conversely, transgenic or pharmacological activation of SHH rescued HFD-induced hair loss. These data collectively demonstrate that stem cell inflammatory signals induced by obesity robustly represses organ regeneration signals to accelerate the miniaturization of mini-organs, and suggests the importance of daily prevention of organ dysfunction.
Collapse
Affiliation(s)
- Hironobu Morinaga
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuaki Mohri
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Marina Grachtchouk
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kyosuke Asakawa
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Matsumura
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Motohiko Oshima
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Naoya Takayama
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoki Kato
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuriko Nishimori
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuriko Sorimachi
- Department of Stem Cell Biology, Research Institute National Center for Global Health and Medicine, Tokyo, Japan
| | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute National Center for Global Health and Medicine, Tokyo, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichiro Iwakura
- Centre for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Andrzej A Dlugosz
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Emi K Nishimura
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
- Division of Aging and Regeneration, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Division of Stem Cell Aging Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|