1
|
Zhang P, Wang J, Miao J, Zhu P. The dual role of tissue regulatory T cells in tissue repair: return to homeostasis or fibrosis. Front Immunol 2025; 16:1560578. [PMID: 40114929 PMCID: PMC11922884 DOI: 10.3389/fimmu.2025.1560578] [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: 01/14/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Tissue resident regulatory T cells (tissue Tregs) are vital for maintaining immune homeostasis and controlling inflammation. They aid in repairing damaged tissues and influencing the progression of fibrosis. However, despite extensive research on how tissue Tregs interact with immune and non-immune cells during tissue repair, their pro- and anti-fibrotic effects in chronic tissue injury remain unclear. Understanding how tissue Tregs interact with various cell types, as well as their roles in chronic injury and fibrosis, is crucial for uncovering the mechanisms behind these conditions. In this review, we describe the roles of tissue Tregs in repair and fibrosis across different tissues and explore potential strategies for regulating tissue homeostasis. These insights hold promise for providing new perspectives and approaches for the treatment of irreversible fibrotic diseases.
Collapse
Affiliation(s)
| | | | - Jinlin Miao
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Ping Zhu
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
2
|
Sugiura S, Yoshida H, Sugiura H, Uehara M, Sugiura Y, Maruo Y, Hayashi Y, Yamamoto T, Kato T, Fujimoto N, Udagawa J. Increased intracellular stress responses and decreased KLF2 in adult patients with atopic dermatitis. Cell Stress Chaperones 2025; 30:84-99. [PMID: 39938773 PMCID: PMC11891603 DOI: 10.1016/j.cstres.2025.02.001] [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: 09/08/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
Atopic dermatitis (AD) is prone to exacerbations in response to various triggering factors and flare-ups after remission. We searched for molecules associated with relapse/exacerbation of AD among molecules with altered gene expression in the skin of patients with AD. Microarray analyses were performed on lesional and nonlesional skin of adolescent or adult patients with recalcitrant AD and healthy controls. Five chaperones involved in intracellular stress responses, namely heat shock protein family A (Hsp70) member 9 (HSPA9), heat shock protein 90 beta family member 1 (HSP90B1), calnexin (CANX), malectin (MLEC; endoplasmic reticulum-associated degradation), and heat shock protein family D (Hsp60) member 1 (HSPD1), were consistently upregulated in involved and uninvolved skin of patients with AD. Damage-associated molecular patterns were upregulated in involved skin. KLF transcription factor 2 (KLF2) was decreased in involved skin and exhibited a decreasing trend in uninvolved skin of patients with AD. CD4(+)/CD8(+) double-positive cells (1.4% of T cells) were detected in lesions with declined KLF2 levels. WNT inhibitory factor 1 (WIF1) was downregulated in involved skin. Prolactin-induced protein was upregulated in only uninvolved skin of patients with AD. We found increased intracellular stress responses and decreased expression of KLF2 in the skin of patients with AD. Multifactorial genetic diseases, such as asthma, inflammatory bowel disease, type 2 diabetes, and rheumatoid arthritis, are associated with intracellular stress. Intracellular abnormalities may also be responsible for AD. Further research on AD may incorporate enhanced intracellular stress response and the decreased expression of KLF2 into the mechanism underlying AD.
Collapse
Affiliation(s)
- Shuji Sugiura
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan; Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan.
| | - Hiderou Yoshida
- Department of Molecular Biochemistry, Graduate School of Life Science, University of Hyogo, Ako, Japan
| | - Hisashi Sugiura
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan; Department of Dermatology, Sugiura Dermatology Clinic, Kusatsu, Japan
| | - Masami Uehara
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Yasuo Sugiura
- International Health Care Center, National Center for Global Health and Medicine, Tokyo, Japan; Department of Pediatrics, Navitas Clinic, Tokyo, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Yuji Hayashi
- Hospital Division of Diagnostic Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Takefumi Yamamoto
- Central Research Laboratory, Shiga University of Medical Science, Otsu, Japan
| | - Takeshi Kato
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Noriki Fujimoto
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Jun Udagawa
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
3
|
Miao W, Jain V, Han M, Jin YJ, Beasley GM, Starczysnowski DT, Gregory SG, Zhang JY. Inhibition of UBE2N in regulatory T-cells boosts immunity against cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619436. [PMID: 39484562 PMCID: PMC11526935 DOI: 10.1101/2024.10.22.619436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Regulatory T (Treg) cells prevent autoimmunity and facilitate cancer immune evasion. Depletion of Tregs is a promising cancer therapy, but risks of autoimmune reactions hamper its clinical translation. Here, we demonstrate that temporally induced deletion of Ube2n in Tregs (Ube2n Treg-KO ) of adult mice results in a robust expansion and activation of cytotoxic CD8 + T-cells in response to cancer cell challenges, producing a long-lasting survival benefit without autoimmune complications. The anti-tumor effect persists following adoptive T-cell transfer to T-cell-deficient Rag1-knockout mice. Single-cell transcriptomic analysis revealed that UBE2N deletion shifted immunosuppressive Tregs to effector-like T-cells. This shift is characterized by the downregulation of c-Myc target genes, resembling that observed in tumor-infiltrating Tregs of melanoma patients. Further analyses confirm that UBE2N maintains c-Myc protein stability via suppression of K48-Ubiquitin-mediated proteasomal degradation. Taken together, our studies uncover a hitherto unexplored and potentially druggable UBE2N/c-Myc signaling axis to eradicate Treg-enabled cancer immune escape.
Collapse
|
4
|
Hossain MM, King P, Hackett J, Gerard HC, Niwinski R, Wu L, Van Kaer L, Dyson G, Gibson H, Borowsky AD, Sebzda E. Peripheral-derived regulatory T cells contribute to tumor-mediated immune suppression in a nonredundant manner. Proc Natl Acad Sci U S A 2024; 121:e2404916121. [PMID: 39207730 PMCID: PMC11388331 DOI: 10.1073/pnas.2404916121] [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: 03/08/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Identifying tumor-mediated mechanisms that impair immunity is instrumental for the design of new cancer therapies. Regulatory T cells (Tregs) are a key component of cancer-derived immune suppression; however, these lymphocytes are necessary to prevent systemic autoimmunity in mice and humans, and thus, direct targeting of Tregs is not a clinical option for cancer patients. We have previously demonstrated that excising transcription factor Kruppel-like factor 2 (Klf2) within the T cell lineage blocks the generation of peripheral-derived Tregs (pTregs) without impairing production of thymic-derived Tregs. Using this mouse model, we have now demonstrated that eliminating pTregs is sufficient to delay/prevent tumor malignancy without causing autoimmunity. Cancer-bearing mice that expressed KLF2 converted tumor-specific CD4+ T cells into pTregs, which accumulated in secondary lymphoid organs and impaired further T cell effector activity. In contrast, pTreg-deficient mice retained cancer-specific immunity, including improved T cell infiltration into "cold" tumors, reduced T cell exhaustion in tumor beds, restricted generation of tumor-associated myeloid-derived suppressor cells, and the continued production of circulating effector T cells that arose in a cancer-dependent manner. Results indicate that tumor-specific pTregs are critical for early stages of cancer progression and blocking the generation of these inhibitory lymphocytes safely delays/prevents malignancy in preclinical models of melanoma and prostate cancer.
Collapse
Affiliation(s)
- Md Moazzem Hossain
- Department of Biochemistry, Microbiology and Immunology, Wayne State University Medical School, Detroit, MI48201
| | - Paul King
- Department of Biochemistry, Microbiology and Immunology, Wayne State University Medical School, Detroit, MI48201
| | - Justin Hackett
- Department of Oncology, Wayne State University Medical School, Detroit, MI48201
| | - Herve C. Gerard
- Department of Biochemistry, Microbiology and Immunology, Wayne State University Medical School, Detroit, MI48201
| | - Rajmund Niwinski
- Department of Biochemistry, Microbiology and Immunology, Wayne State University Medical School, Detroit, MI48201
| | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN37232
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN37232
| | - Gregory Dyson
- Department of Oncology, Wayne State University Medical School, Detroit, MI48201
- Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI48201
| | - Heather Gibson
- Department of Biochemistry, Microbiology and Immunology, Wayne State University Medical School, Detroit, MI48201
- Department of Oncology, Wayne State University Medical School, Detroit, MI48201
- Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI48201
| | - Alexander D. Borowsky
- Department of Pathology and Laboratory Medicine, Center for Comparative Medicine, University of California Davis, Davis, CA95616
| | - Eric Sebzda
- Department of Biochemistry, Microbiology and Immunology, Wayne State University Medical School, Detroit, MI48201
- Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI48201
| |
Collapse
|
5
|
Wang Z, Lang T, Li Y, Zhang X, Abdur M, Mao M. Hypermethylation of the FOXP3 gene regulates Tregs immunodysregulation in chronic idiopathic thrombocytopenic purpura. Allergol Immunopathol (Madr) 2024; 52:30-37. [PMID: 38970262 DOI: 10.15586/aei.v52i4.1091] [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: 02/23/2024] [Accepted: 04/29/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Chronic idiopathic thrombocytopenic purpura (ITP) is an autoimmune disease characterized by a breakdown of immune tolerance; in ITP, the body's immune system mistakenly attacks and destroys platelets. This study aims to investigate the role and underlying mechanisms of FOXP3 in chronic ITP. METHODS Flow cytometry was used to detect the proportion of CD4+CD25+FOXP3+ regulatory T cells (Tregs) in CD4+CD25+ T lymphocytes from 20 patients with chronic ITP (CITP), 20 acute ITP (AITP) controls, and 20 healthy individuals.CD4+CD25+ Treg cells were isolated from peripheral blood of patients with CITP using magnetic beads and then treated with phosphate-buffered saline solution or decitabine (a methylation inhibitor) for 48 h. The levels of interleukin-2 (IL-2), IL-10, and transforming growth factor-beta1 (TGF-β1) in the plasma and CD4+CD25+ Treg cells were assessed by Enzyme-linked-immunosorbent serologic assay and quantitative real-time polymerase chain reaction (qRT-PCR). FOXP3 level was measured by qRT-PCR and Western blot analysis. Methylation-specific PCR (MS-PCR) was adopted to detect the status of FOXP3 methylation. RESULTS The number of Treg cells and the contents of IL-2, IL-10, and TGF-β1 decreased in patients with CITP, compared to the AITP control group and normal group. FOXP3 expression was reduced and FOXP3 methylation increased in patients with CITP, compared to the AITP control group and normal group. Hypermethylation of FOXP3 promoter led to decrease in FOXP3 level in Treg cells. Inhibition of FOXP3 promoter hypermethylation promoted the secretion of IL-2, IL-10, and TGF-β1 in Treg cells. CONCLUSION The number of Treg cells in CITP patients decreased, and the hypermethylation of FOXP3 promoter led to reduction of its expression in Treg cells, thus affecting the immune functioning of Treg cells.
Collapse
Affiliation(s)
- Zengsheng Wang
- Department of Hematology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Tao Lang
- Department of Hematology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Yan Li
- Department of Hematology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xiaoyan Zhang
- Department of Hematology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Muhubair Abdur
- Department of Hematology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China;
| | - Min Mao
- Department of Hematology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
6
|
Shao TY, Jiang TT, Stevens J, Russi AE, Troutman TD, Bernieh A, Pham G, Erickson JJ, Eshleman EM, Alenghat T, Jameson SC, Hogquist KA, Weaver CT, Haslam DB, Deshmukh H, Way SS. Kruppel-like factor 2+ CD4 T cells avert microbiota-induced intestinal inflammation. Cell Rep 2023; 42:113323. [PMID: 37889750 PMCID: PMC10822050 DOI: 10.1016/j.celrep.2023.113323] [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: 12/12/2022] [Revised: 09/05/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Intestinal colonization by antigenically foreign microbes necessitates expanded peripheral immune tolerance. Here we show commensal microbiota prime expansion of CD4 T cells unified by the Kruppel-like factor 2 (KLF2) transcriptional regulator and an essential role for KLF2+ CD4 cells in averting microbiota-driven intestinal inflammation. CD4 cells with commensal specificity in secondary lymphoid organs and intestinal tissues are enriched for KLF2 expression, and distinct from FOXP3+ regulatory T cells or other differentiation lineages. Mice with conditional KLF2 deficiency in T cells develop spontaneous rectal prolapse and intestinal inflammation, phenotypes overturned by eliminating microbiota or reconstituting with donor KLF2+ cells. Activated KLF2+ cells selectively produce IL-10, and eliminating IL-10 overrides their suppressive function in vitro and protection against intestinal inflammation in vivo. Together with reduced KLF2+ CD4 cell accumulation in Crohn's disease, a necessity for the KLF2+ subpopulation of T regulatory type 1 (Tr1) cells in sustaining commensal tolerance is demonstrated.
Collapse
Affiliation(s)
- Tzu-Yu Shao
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Tony T Jiang
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Joseph Stevens
- Division of Neonatology and Pulmonary Biology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Abigail E Russi
- Division of Gastroenterology, Hepatology and Advanced Nutrition, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Ty D Troutman
- Division of Allergy and Immunology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Anas Bernieh
- Division of Pathology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Giang Pham
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - John J Erickson
- Division of Neonatology and Pulmonary Biology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Emily M Eshleman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Theresa Alenghat
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Kristin A Hogquist
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Casey T Weaver
- Program in Immunology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - David B Haslam
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Hitesh Deshmukh
- Division of Neonatology and Pulmonary Biology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
7
|
Luckner B, Essfeld F, Ayobahan SU, Richling E, Eilebrecht E, Eilebrecht S. Transcriptomic profiling of TLR-7-mediated immune-challenge in zebrafish embryos in the presence and absence of glucocorticoid-induced immunosuppression. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115570. [PMID: 37844410 DOI: 10.1016/j.ecoenv.2023.115570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/28/2023] [Accepted: 10/08/2023] [Indexed: 10/18/2023]
Abstract
Although numerous studies imply a correlation between chemical contamination and an impaired immunocompetence of wildlife populations, the assessment of immunomodulatory modes of action is currently not covered in the regulatory requirements for the approval of new substances. This is not least due to the complexity of the immune system and a lack of standardised methods and validated biomarkers. To tackle this issue, in this study, the transcriptomic profiles of zebrafish embryos were analysed in response to the immunosuppressive compound clobetasol propionate, a synthetic glucocorticoid, and/or the immunostimulatory compound imiquimod (IMQ), a TLR-7 agonist. Using IMQ, known for its potential to induce psoriasis-like effects in mice and human, this study additionally aimed at evaluating the usability of the zebrafish embryo model as an alternative and 3R conform system for the IMQ-induced psoriasis mouse model. Our study substantiates the suitability of previously proposed genes as possible biomarkers for immunotoxicity, such as socs3, nfkbia, anxa1c, fkbp5 and irg1l. Likewise, however, our findings indicate that these genes may be less suitable to distinguish a suppressive from stimulating fashion of action. In contrast, based on a differential regulation in opposite direction in response to both compounds, krt17, rtn4a, and1, smhyc1 and gmpr were identified as potential novel biomarkers with said power to differentiate. Observed IMQ-induced alterations in the expression of genes previously associated with the pathogenesis of psoriasis such as krt17, nfkbia, parp1, pparg, nfil3-6, per2, stat4, klf2, rtn4a, anxa1c and nr1d2 indicate the inducibility of psoriatic effects in the zebrafish embryo. Our work contributes to the establishment of an approach for a 3R-compliant investigation of immunotoxic mechanisms of action in aquatic vertebrates. The validated and newly identified biomarker candidates of specific immunotoxic effects can be used in future studies in the context of environmental hazard assessment of substances or also for human-relevant immunotoxicological questions.
Collapse
Affiliation(s)
- Benedikt Luckner
- Department Ecotoxicogenomics, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Schmallenberg, Germany
| | - Fabian Essfeld
- Department Ecotoxicogenomics, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Schmallenberg, Germany; Computational Biology, Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Steve U Ayobahan
- Department Ecotoxicogenomics, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Schmallenberg, Germany
| | - Elke Richling
- Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, Germany
| | - Elke Eilebrecht
- Department Ecotoxicology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Schmallenberg, Germany
| | - Sebastian Eilebrecht
- Department Ecotoxicogenomics, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Schmallenberg, Germany.
| |
Collapse
|
8
|
Larson JH, Jin S, Loschi M, Bolivar Wagers S, Thangavelu G, Zaiken MC, McDonald-Hyman C, Saha A, Aguilar EG, Koehn B, Osborn MJ, Panoskaltsis-Mortari A, Macdonald KPA, Hill GR, Murphy WJ, Serody JS, Maillard I, Kean LS, Kim SV, Littman DR, Blazar BR. Enforced gut homing of murine regulatory T cells reduces early graft-versus-host disease severity. Am J Transplant 2023; 23:1102-1115. [PMID: 36878433 PMCID: PMC10475494 DOI: 10.1016/j.ajt.2023.01.030] [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: 12/19/2022] [Accepted: 01/31/2023] [Indexed: 03/07/2023]
Abstract
Damage to the gastrointestinal tract following allogeneic hematopoietic stem cell transplantation is a significant contributor to the severity and perpetuation of graft-versus-host disease. In preclinical models and clinical trials, we showed that infusing high numbers of regulatory T cells reduces graft-versus-host disease incidence. Despite no change in in vitro suppressive function, transfer of ex vivo expanded regulatory T cells transduced to overexpress G protein-coupled receptor 15 or C-C motif chemokine receptor 9, specific homing receptors for colon or small intestine, respectively, lessened graft-versus-host disease severity in mice. Increased regulatory T cell frequency and retention within the gastrointestinal tissues of mice that received gut homing T cells correlated with lower inflammation and gut damage early post-transplant, decreased graft-versus-host disease severity, and prolonged survival compared with those receiving control transduced regulatory T cells. These data provide evidence that enforced targeting of ex vivo expanded regulatory T cells to the gastrointestinal tract diminishes gut injury and is associated with decreased graft-versus-host disease severity.
Collapse
Affiliation(s)
- Jemma H Larson
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sujeong Jin
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael Loschi
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sara Bolivar Wagers
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Govindarajan Thangavelu
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael C Zaiken
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cameron McDonald-Hyman
- Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Asim Saha
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ethan G Aguilar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brent Koehn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark J Osborn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kelli P A Macdonald
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Immunology Department, Brisbane, Queensland, Australia
| | - Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA; Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| | - William J Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA; Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, California, USA
| | - Jonathan S Serody
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sangwon V Kim
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Dan R Littman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, USA; Howard Hughes Medical Institute, New York University School of Medicine, New York, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
9
|
Cassidy MF, Herbert ZT, Moulton VR. Splicing factor SRSF1 controls autoimmune-related molecular pathways in regulatory T cells distinct from FoxP3. Mol Immunol 2022; 152:140-152. [PMID: 36368121 DOI: 10.1016/j.molimm.2022.10.017] [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: 01/18/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
Regulatory T cells (Tregs) are vital for maintaining immune self-tolerance, and their impaired function leads to autoimmune disease. Mutations in FoxP3, the master transcriptional regulator of Tregs, leads to immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome in humans and the early lethal "scurfy" phenotype with multi-organ autoimmune disease in mice. We recently identified serine/arginine-rich splicing factor 1 (SRSF1) as an indispensable regulator of Treg homeostasis and function. Intriguingly, Treg-conditional SRSF1-deficient mice exhibit early lethal systemic autoimmunity with multi-organ inflammation reminiscent of the scurfy mice. Importantly, SRSF1 is decreased in T cells from patients with the autoimmune disease systemic lupus erythematosus (SLE), and low SRSF1 levels inversely correlate with disease severity. Given that the Treg-specific deficiency of SRSF1 causes similarly profound autoimmune disease outcomes in mice as the deficiency/mutation in FoxP3, we aimed to evaluate the genes and molecular pathways controlled by these two indispensable regulatory proteins. We performed comparative bioinformatic analyses of transcriptomic profiles of Tregs from Srsf1-knockout mice and two Foxp3 mutant mice--the FoxP3-deficient ΔFoxp3 and the Foxp3 M370I mutant mice. We identified 132 differentially expressed genes (DEGs) unique to Srsf1-ko Tregs, 503 DEGs unique to Foxp3 M370I Tregs, and 1367 DEGs unique to ΔFoxp3 Tregs. Gene set enrichment and pathway analysis of DEGs unique to Srsf1-ko Tregs indicate that SRSF1 controls cytokine and immune response pathways. Conversely, FoxP3 controls pathways involved in DNA replication and cell cycle. Besides the distinct gene signatures, we identified only 30 shared genes between all three Treg mutants, mostly contributing to cytokine and immune defense pathways. Prominent genes included the chemokines CXCR6 and CCL1 and the checkpoint inhibitors FASLG and PDCD1. Thus, we demonstrate that SRSF1 and FoxP3 control common and distinct molecular pathways implicated in autoimmunity. Our analyses suggest that SRSF1 controls crucial immune functions in Tregs contributing to immune tolerance, and perturbations in its levels lead to systemic autoimmunity via mechanisms that are largely distinct from FoxP3.
Collapse
Affiliation(s)
- Michael F Cassidy
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Tufts University School of Medicine, Boston, MA, United States.
| | - Zachary T Herbert
- Molecular Biology Core Facilities, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Vaishali R Moulton
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Pernaa N, Keskitalo S, Chowdhury I, Nissinen A, Glumoff V, Keski-Filppula R, Junttila J, Eklund KK, Santaniemi W, Siitonen S, Seppänen MRJ, Vähäsalo P, Varjosalo M, Åström P, Hautala T. Heterozygous premature termination in zinc-finger domain of Krüppel-like factor 2 gene associates with dysregulated immunity. Front Immunol 2022; 13:819929. [PMID: 36466816 PMCID: PMC9716311 DOI: 10.3389/fimmu.2022.819929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 10/21/2022] [Indexed: 08/23/2024] Open
Abstract
Krüppel-like factor 2 (KLF2) is a transcription factor with significant roles in development, maturation, differentiation, and proliferation of several cell types. In immune cells, KLF2 regulates maturation and trafficking of lymphocytes and monocytes. KLF2 participates in regulation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. Although pulmonary arterial hypertension (PAH) related to KLF2 genetic variant has been suggested, genetic role of KLF2 associated with immune dysregulation has not been described. We identified a family whose members suffered from lymphopenia, autoimmunity, and malignancy. Whole exome sequencing revealed a KLF2 p.(Glu318Argfs*87) mutation disrupting the highly conserved zinc finger domain. We show a reduced amount of KLF2 protein, defective nuclear localization and altered protein-protein interactome. The phenotypically variable positive cases presented with B and T cell lymphopenia and abnormalities in B and T cell maturation including low naive T cell counts and low CD27+IgD-IgM- switched memory B cells. KLF2 target gene (CD62L) expression was affected. Although the percentage of (CD25+FOXP3+, CD25+CD127-) regulatory T cells (Treg) was high, the naive Treg cells (CD45RA+) were absent. Serum IgG1 levels were low and findings in one case were consistent with common variable immunodeficiency (CVID). Transcription of NF-κβ pathway genes and p65/RelA phosphorylation were not significantly affected. Inflammasome activity, transcription of genes related with JAK/STAT pathway and interferon signature were also comparable to controls. Evidence of PAH was not found. In conclusion, KLF2 variant may be associated with familial immune dysregulation. Although the KLF2 deficient family members in our study suffered from lymphopenia, autoimmunity or malignancy, additional study cohorts are required to confirm our observations.
Collapse
Affiliation(s)
- Nora Pernaa
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Salla Keskitalo
- Molecular Systems Biology Group, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Iftekhar Chowdhury
- Molecular Systems Biology Group, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Antti Nissinen
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Virpi Glumoff
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Riikka Keski-Filppula
- PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Department of Clinical Genetics, Oulu University Hospital, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Juhani Junttila
- Research Unit of Internal Medicine, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Kari K. Eklund
- Department of Rheumatology, Inflammation Center, University of Helsinki and Helsinki University Hospital and Orton Orthopedic Hospital, Helsinki, Finland
| | - Wenny Santaniemi
- Oulun University Hospital and Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Sanna Siitonen
- Department of Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Mikko RJ. Seppänen
- Rare Disease Center and Pediatric Research Center, Children and Adolescents; Adult Immunodeficiency Unit, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Paula Vähäsalo
- PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Department of Pediatrics, Oulu University Hospital, Oulu, Finland
| | - Markku Varjosalo
- Molecular Systems Biology Group, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Pirjo Åström
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Timo Hautala
- Infectious Diseases, Oulu University Hospital and Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| |
Collapse
|
11
|
Guo H, Wang M, Wang B, Guo L, Cheng Y, Wang Z, Sun YQ, Wang Y, Chang YJ, Huang XJ. PRDM1 Drives Human Primary T Cell Hyporesponsiveness by Altering the T Cell Transcriptome and Epigenome. Front Immunol 2022; 13:879501. [PMID: 35572579 PMCID: PMC9097451 DOI: 10.3389/fimmu.2022.879501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022] Open
Abstract
T cell hyporesponsiveness is crucial for the functional immune system and prevents the damage induced by alloreactive T cells in autoimmune pathology and transplantation. Here, we found low expression of PRDM1 in T cells from donor and recipients both related to the occurrence of acute graft-versus-host disease (aGVHD). Our systematic multiomics analysis found that the transcription factor PRDM1 acts as a master regulator during inducing human primary T cell hyporesponsiveness. PRDM1-overexpression in primary T cells expanded Treg cell subset and increased the expression level of FOXP3, while decreased expression had the opposite effects. Moreover, the binding motifs of key T cell function regulators, such as FOS, JUN and AP-1, were enriched in PRDM1 binding sites and that PRDM1 altered the chromatin accessibility of these regions. Multiomics analysis showed that PRDM1 directly upregulated T cell inhibitory genes such as KLF2 and KLRD1 and downregulated the T cell activation gene IL2, indicating that PRDM1 could promote a tolerant transcriptional profile. Further analysis showed that PRDM1 upregulated FOXP3 expression level directly by binding to FOXP3 upstream enhancer region and indirectly by upregulating KLF2. These results indicated that PRDM1 is sufficient for inducing human primary T cell hyporesponsiveness by transcriptomic and epigenetic manners.
Collapse
Affiliation(s)
- Huidong Guo
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ming Wang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bixia Wang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liping Guo
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yifei Cheng
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Zhidong Wang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu-Qian Sun
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu Wang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China.,Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Peking University, Beijing, China.,Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies (2019RU029), Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Eder JM, Sacco RE. Ex vivo activated CD4+ T cells from young calves exhibit Th2-biased effector function with distinct metabolic reprogramming compared to adult cows. Vet Immunol Immunopathol 2022; 248:110418. [DOI: 10.1016/j.vetimm.2022.110418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/29/2022] [Accepted: 04/02/2022] [Indexed: 10/18/2022]
|
13
|
Wittner J, Schulz SR, Steinmetz TD, Berges J, Hauke M, Channell WM, Cunningham AF, Hauser AE, Hutloff A, Mielenz D, Jäck HM, Schuh W. Krüppel-like factor 2 controls IgA plasma cell compartmentalization and IgA responses. Mucosal Immunol 2022; 15:668-682. [PMID: 35347229 PMCID: PMC9259478 DOI: 10.1038/s41385-022-00503-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/24/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023]
Abstract
Krüppel-like factor 2 (KLF2) is a potent regulator of lymphocyte differentiation, activation and migration. However, its functional role in adaptive and humoral immunity remains elusive. Therefore, by using mice with a B cell-specific deletion of KLF2, we investigated plasma cell differentiation and antibody responses. We revealed that the deletion of KLF2 resulted in perturbed IgA plasma cell compartmentalization, characterized by the absence of IgA plasma cells in the bone marrow, their reductions in the spleen, the blood and the lamina propria of the colon and the small intestine, concomitant with their accumulation and retention in mesenteric lymph nodes and Peyer's patches. Most intriguingly, secretory IgA in the intestinal lumen was almost absent, dimeric serum IgA was drastically reduced and antigen-specific IgA responses to soluble Salmonella flagellin were blunted in KLF2-deficient mice. Perturbance of IgA plasma cell localization was caused by deregulation of CCR9, Integrin chains αM, α4, β7, and sphingosine-1-phosphate receptors. Hence, KLF2 not only orchestrates the localization of IgA plasma cells by fine-tuning chemokine receptors and adhesion molecules but also controls IgA responses to Salmonella flagellin.
Collapse
Affiliation(s)
- Jens Wittner
- grid.411668.c0000 0000 9935 6525Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian R. Schulz
- grid.411668.c0000 0000 9935 6525Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Tobit D. Steinmetz
- grid.411668.c0000 0000 9935 6525Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Berges
- grid.411668.c0000 0000 9935 6525Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Manuela Hauke
- grid.411668.c0000 0000 9935 6525Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - William M. Channell
- grid.6572.60000 0004 1936 7486Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Adam F. Cunningham
- grid.6572.60000 0004 1936 7486Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Anja E. Hauser
- grid.6363.00000 0001 2218 4662Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany ,grid.418217.90000 0000 9323 8675Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Andreas Hutloff
- grid.412468.d0000 0004 0646 2097Institute of Immunology and Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Dirk Mielenz
- grid.411668.c0000 0000 9935 6525Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- grid.411668.c0000 0000 9935 6525Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Schuh
- grid.411668.c0000 0000 9935 6525Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger Center, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
14
|
Krüppel-like Factor 2 (KLF2) in Immune Cell Migration. Vaccines (Basel) 2021; 9:vaccines9101171. [PMID: 34696279 PMCID: PMC8539188 DOI: 10.3390/vaccines9101171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 01/30/2023] Open
Abstract
Krüppel-like factor 2 (KLF2), a transcription factor of the krüppel-like family, is a key regulator of activation, differentiation, and migration processes in various cell types. In this review, we focus on the functional relevance of KLF2 in immune cell migration and homing. We summarize the key functions of KLF2 in the regulation of chemokine receptors and adhesion molecules and discuss the relevance of the KLF2-mediated control of immune cell migration in the context of immune responses, infections, and diseases.
Collapse
|
15
|
Song Q, Lei Y, Shao L, Li W, Kong Q, Lin Z, Qin X, Wei W, Hou F, Li J, Guo X, Mao Y, Cao Y, Liu Z, Zheng L, Liang R, Jiang Y, Liu Y, Zhang L, Yang J, Lau YL, Zhang Y, Ban B, Wang YF, Yang W. Genome-wide association study on Northern Chinese identifies KLF2, DOT1L and STAB2 associated with systemic lupus erythematosus. Rheumatology (Oxford) 2021; 60:4407-4417. [PMID: 33493351 DOI: 10.1093/rheumatology/keab016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/10/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To identify novel genetic loci associated with systemic lupus erythematosus (SLE) and to evaluate potential genetic differences between ethnic Chinese and European populations in SLE susceptibility. METHODS A new genome-wide association study (GWAS) was conducted from Jining, North China, on 1506 individuals (512 SLE cases and 994 matched healthy controls). The association results were meta-analysed with existing data on Chinese populations from Hong Kong, Guangzhou and Central China, as well as GWAS results from four cohorts of European ancestry. A total of 26 774 individuals (9310 SLE cases and 17 464 controls) were included in this study. RESULTS Meta-analysis on four Chinese cohorts identifies KLF2 as a novel locus associated with SLE [rs2362475; odds ratio (OR) = 0.85, P=2.00E-09]. KLF2 is likely an Asian-specific locus as no evidence of association was detected in the four European cohorts (OR = 0.98, P =0.58), with evidence of heterogeneity (P=0.0019) between the two ancestral groups. Meta-analyses of results from both Chinese and Europeans identify STAB2 (rs10082873; OR= 0.89, P=4.08E-08) and DOT1L (rs4807205; OR= 1.12, P=8.17E-09) as trans-ancestral association loci, surpassing the genome-wide significance. CONCLUSIONS We identified three loci associated with SLE, with KLF2 a likely Chinese-specific locus, highlighting the importance of studying diverse populations in SLE genetics. We hypothesize that DOT1L and KLF2 are plausible SLE treatment targets, with inhibitors of DOT1L and inducers of KLF2 already available clinically.
Collapse
Affiliation(s)
- Qin Song
- Department of Rheumatology and Lupus Research Institute, The Affiliated Hospital of Jining Medical University
| | - Yao Lei
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Shandong
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Li Shao
- Department of Rheumatology and Lupus Research Institute, The Affiliated Hospital of Jining Medical University
| | - Weiyang Li
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Shandong
| | - Qingsheng Kong
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Shandong
| | - Zhiming Lin
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University
| | - Xiao Qin
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Shandong
| | - Wei Wei
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Shandong
| | - Fei Hou
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Shandong
| | - Jian Li
- Department of Rheumatology and Lupus Research Institute, The Affiliated Hospital of Jining Medical University
| | - Xianghua Guo
- Department of Rheumatology and Lupus Research Institute, The Affiliated Hospital of Jining Medical University
| | - Yujing Mao
- Department of Rheumatology and Lupus Research Institute, The Affiliated Hospital of Jining Medical University
| | - Yujie Cao
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Zhongyi Liu
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Lichuan Zheng
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Rui Liang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Yuping Jiang
- Department of Rheumatology and Lupus Research Institute, The Affiliated Hospital of Jining Medical University
| | - Yan Liu
- Department of Rheumatology and Lupus Research Institute, The Affiliated Hospital of Jining Medical University
| | - Lili Zhang
- Department of Rheumatology and Lupus Research Institute, The Affiliated Hospital of Jining Medical University
| | - Jing Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Yan Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou
| | - Bo Ban
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining Medical University
- Chinese Research Center for Behavior Medicine in Growth and Development, Shandong
| | - Yong-Fei Wang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| |
Collapse
|
16
|
TCF-1 controls T reg cell functions that regulate inflammation, CD8 + T cell cytotoxicity and severity of colon cancer. Nat Immunol 2021; 22:1152-1162. [PMID: 34385712 PMCID: PMC8428683 DOI: 10.1038/s41590-021-00987-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
The transcription factor TCF-1 is essential for the development and function of regulatory T (Treg) cells; however, its function is poorly understood. Here, we show that TCF-1 primarily suppresses transcription of genes that are co-bound by Foxp3. Single-cell RNA-sequencing analysis identified effector memory T cells and central memory Treg cells with differential expression of Klf2 and memory and activation markers. TCF-1 deficiency did not change the core Treg cell transcriptional signature, but promoted alternative signaling pathways whereby Treg cells became activated and gained gut-homing properties and characteristics of the TH17 subset of helper T cells. TCF-1-deficient Treg cells strongly suppressed T cell proliferation and cytotoxicity, but were compromised in controlling CD4+ T cell polarization and inflammation. In mice with polyposis, Treg cell-specific TCF-1 deficiency promoted tumor growth. Consistently, tumor-infiltrating Treg cells of patients with colorectal cancer showed lower TCF-1 expression and increased TH17 expression signatures compared to adjacent normal tissue and circulating T cells. Thus, Treg cell-specific TCF-1 expression differentially regulates TH17-mediated inflammation and T cell cytotoxicity, and can determine colorectal cancer outcome.
Collapse
|
17
|
Shahneh F, Grill A, Klein M, Frauhammer F, Bopp T, Schäfer K, Raker VK, Becker C. Specialized regulatory T cells control venous blood clot resolution through SPARC. Blood 2021; 137:1517-1526. [PMID: 32932520 DOI: 10.1182/blood.2020005407] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
The cells and mechanisms involved in blood clot resorption are only partially known. We show that regulatory T cells (Tregs) accumulate in venous blood clots and regulate thrombolysis by controlling the recruitment, differentiation and matrix metalloproteinase (MMP) activity of monocytes. We describe a clot Treg population that forms the matricellular acid- and cysteine-rich protein SPARC (secreted protein acidic and rich in cysteine) and show that SPARC enhances monocyte MMP activity and that SPARC+ Tregs are crucial for blood clot resorption. By comparing different treatment times, we define a therapeutic window of Treg expansion that accelerates clot resorption.
Collapse
Affiliation(s)
- Fatemeh Shahneh
- Department of Dermatology
- Center for Thrombosis and Hemostasis, and
| | | | - Matthias Klein
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany; and
| | - Felix Frauhammer
- Center for Molecular Biology, University of Heidelberg, Heidelberg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany; and
| | | | - Verena K Raker
- Department of Dermatology
- Center for Thrombosis and Hemostasis, and
| | - Christian Becker
- Department of Dermatology
- Center for Thrombosis and Hemostasis, and
| |
Collapse
|
18
|
Permanyer M, Bošnjak B, Glage S, Friedrichsen M, Floess S, Huehn J, Patzer GE, Odak I, Eckert N, Zargari R, Ospina-Quintero L, Georgiev H, Förster R. Efficient IL-2R signaling differentially affects the stability, function, and composition of the regulatory T-cell pool. Cell Mol Immunol 2021; 18:398-414. [PMID: 33408345 PMCID: PMC8027001 DOI: 10.1038/s41423-020-00599-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/14/2020] [Indexed: 01/10/2023] Open
Abstract
Signaling via interleukin-2 receptor (IL-2R) is a requisite for regulatory T (Treg) cell identity and function. However, it is not completely understood to what degree IL-2R signaling is required for Treg cell homeostasis, lineage stability and function in both resting and inflammatory conditions. Here, we characterized a spontaneous mutant mouse strain endowed with a hypomorphic Tyr129His variant of CD25, the α-chain of IL-2R, which resulted in diminished receptor expression and reduced IL-2R signaling. Under noninflammatory conditions, Cd25Y129H mice harbored substantially lower numbers of peripheral Treg cells with stable Foxp3 expression that prevented the development of spontaneous autoimmune disease. In contrast, Cd25Y129H Treg cells failed to efficiently induce immune suppression and lost lineage commitment in a T-cell transfer colitis model, indicating that unimpaired IL-2R signaling is critical for Treg cell function in inflammatory environments. Moreover, single-cell RNA sequencing of Treg cells revealed that impaired IL-2R signaling profoundly affected the balance of central and effector Treg cell subsets. Thus, partial loss of IL-2R signaling differentially interferes with the maintenance, heterogeneity, and suppressive function of the Treg cell pool.
Collapse
Affiliation(s)
- Marc Permanyer
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Silke Glage
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | | | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany
| | | | - Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Nadine Eckert
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Razieh Zargari
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Hristo Georgiev
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
19
|
Fluvastatin-Pretreated Donor Cells Attenuated Murine aGVHD by Balancing Effector T Cell Distribution and Function under the Regulation of KLF2. BIOMED RESEARCH INTERNATIONAL 2021; 2020:7619849. [PMID: 33415155 PMCID: PMC7769635 DOI: 10.1155/2020/7619849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/17/2020] [Accepted: 12/01/2020] [Indexed: 02/04/2023]
Abstract
Prevention of acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is still to be explored. Statins are potent immunomodulatory agents that hold promise as novel and safe agents for aGVHD prophylaxis, yet the controversial effect and regulatory mechanism are incompletely understood. Here, in an MHC mismatched murine model, we found that Fluvastatin-pretreated donor cells could attenuate aGVHD severity by remission tissue pathological injury. Fluvastatin served to restrain effector T cells entry into aGVHD target organs from secondary lymphoid organs (SLOs). The potential mechanism of correcting the effector T cell biased distribution was that Fluvastatin elevated CD62L and CCR7 expression while decreased CXCR3 and CD44 levels, which were correlated with Kruppel-like factor 2 (KLF2) sustention in donor-derived cells. In addition, Fluvastatin was contributed to reducing cytokines IFN-γ, TNF-α, and granzyme-B production in allogeneic effector CD4+ and CD8+ T cells. Furthermore, evidence confirmed that Fluvastatin had a long-lasting effect to sustain KLF2 expression both in vitro and in vivo even under the stimulated circumstance. In conclusion, administration of Fluvastatin to donor mice showed protective effects against recipient aGVHD when compared to untreated mice due to the retention of effector T cells in lymphoid organs accompanying with reduction of nonlymphatic infiltration and related inflammatory cytokines.
Collapse
|
20
|
Vallion R, Divoux J, Glauzy S, Ronin E, Lombardi Y, Lubrano di Ricco M, Grégoire S, Nemazanyy I, Durand A, Fradin D, Lucas B, Salomon BL. Regulatory T Cell Stability and Migration Are Dependent on mTOR. THE JOURNAL OF IMMUNOLOGY 2020; 205:1799-1809. [PMID: 32839235 DOI: 10.4049/jimmunol.1901480] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 07/24/2020] [Indexed: 02/06/2023]
Abstract
CD4+ Foxp3+ regulatory T cells (Treg) are essential to maintain immune tolerance, as their loss leads to a fatal autoimmune syndrome in mice and humans. Conflicting findings have been reported concerning their metabolism. Some reports found that Treg have low mechanistic target of rapamycin (mTOR) activity and would be less dependent on this kinase compared with conventional T cells, whereas other reports suggest quite the opposite. In this study, we revisited this question by using mice that have a specific deletion of mTOR in Treg. These mice spontaneously develop a severe and systemic inflammation. We show that mTOR expression by Treg is critical for their differentiation into effector Treg and their migration into nonlymphoid tissues. We also reveal that mTOR-deficient Treg have reduced stability. This loss of Foxp3 expression is associated with partial Foxp3 DNA remethylation, which may be due to an increased activity of the glutaminolysis pathway. Thus, our work shows that mTOR is crucial for Treg differentiation, migration, and identity and that drugs targeting this metabolism pathway will impact on their biology.
Collapse
Affiliation(s)
- Romain Vallion
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Jordane Divoux
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Salomé Glauzy
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Emilie Ronin
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Yannis Lombardi
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Martina Lubrano di Ricco
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Sylvie Grégoire
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Ivan Nemazanyy
- Plateforme Etude du Métabolisme, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS 3633, Paris, France
| | - Aurélie Durand
- Paris Descartes Université, Sorbonne Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, 75014 Paris, France; and
| | - Delphine Fradin
- CRCINA, Institut de Recherche en Santé de l'Université de Nantes, 44007 Nantes, France
| | - Bruno Lucas
- Paris Descartes Université, Sorbonne Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, 75014 Paris, France; and
| | - Benoit L Salomon
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, CNRS, 75013 Paris, France;
| |
Collapse
|
21
|
Li JQ, Tian JM, Fan XR, Wang ZY, Ling J, Wu XF, Yang FY, Xia YL. miR-106b-5p induces immune imbalance of Treg/Th17 in immune thrombocytopenic purpura through NR4A3/Foxp3 pathway. Cell Cycle 2020; 19:1265-1274. [PMID: 32323598 DOI: 10.1080/15384101.2020.1746485] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Immune imbalance of regulatory T cells (Treg)/T helper 17 cells (Th17) contributes to the development of immune thrombocytopenic purpura (ITP). The dysregulation of miRNAs is important in the development of ITP. However, the role of miR-106b-5p in Treg/Th17 imbalance remains unknown in ITP. MATERIALS AND METHODS Peripheral blood was collected from patients with ITP and healthy controls, and CD4 + T cells were further isolated. miR-106b-5p, nuclear receptor subfamily 4 group A member 3 (NR4A3), forkhead box protein 3 (Foxp3), IL-17A, and TGF-β expressions were detected by qRT-PCR, western blot, or ELISA. The effect of miR-106b-5p on NR4A3 was detected by dual-luciferase reporter gene assay. RESULTS Compared with healthy controls, miR-106b-5p was elevated in peripheral blood of patients with ITP, and NR4A3 expression was decreased. sh-NR4A3 significantly decreased Foxp3 and TGF-β expressions, indicating that NR4A3 may regulate Treg differentiation via Foxp3. Additionally, NR4A3 was identified to be a target of miR-106b-5p, and miR-106b-5p was able to negatively modulate NR4A3 expression. Moreover, we found miR-106b-5p induced immune imbalance of Treg/Th17 through NR4A3. In vivo experiments revealed that silencing miR-106b-5p promoted Treg differentiation and increased the number of platelets, suggesting the relief of ITP. CONCLUSION miR-106b-5p regulated immune imbalance of Treg/Th17 in ITP through the NR4A3/Foxp3 pathway.
Collapse
Affiliation(s)
- Jian-Qin Li
- Department of Hematology, Children's Hospital of Soochow University , Soochow, Jiangsu, China
| | - Jian-Mei Tian
- Department of Hematology, Children's Hospital of Soochow University , Soochow, Jiangsu, China
| | - Xiao-Ru Fan
- Department of Hematology, Children's Hospital of Soochow University , Soochow, Jiangsu, China
| | - Zhao-Yue Wang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University , Soochow, Jiangsu, China
| | - Jing Ling
- Department of Hematology, Children's Hospital of Soochow University , Soochow, Jiangsu, China
| | - Xiao-Fang Wu
- Department of Hematology, Children's Hospital of Soochow University , Soochow, Jiangsu, China
| | - Fei-Yun Yang
- Department of Hematology, Children's Hospital of Soochow University , Soochow, Jiangsu, China
| | - Ya-Lin Xia
- Department of Hematology, Children's Hospital of Soochow University , Soochow, Jiangsu, China
| |
Collapse
|
22
|
Lu DR, Wu H, Driver I, Ingersoll S, Sohn S, Wang S, Li CM, Phee H. Dynamic changes in the regulatory T-cell heterogeneity and function by murine IL-2 mutein. Life Sci Alliance 2020; 3:3/5/e201900520. [PMID: 32269069 PMCID: PMC7156283 DOI: 10.26508/lsa.201900520] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/30/2022] Open
Abstract
Single-cell RNA-seq analysis reveals that IL-2 mutein treatment expands multiple sub-states of regulatory T cells with superior suppressive function in mice. The therapeutic expansion of Foxp3+ regulatory T cells (Tregs) shows promise for treating autoimmune and inflammatory disorders. Yet, how this treatment affects the heterogeneity and function of Tregs is not clear. Using single-cell RNA-seq analysis, we characterized 31,908 Tregs from the mice treated with a half-life extended mutant form of murine IL-2 (IL-2 mutein, IL-2M) that preferentially expanded Tregs, or mouse IgG Fc as a control. Cell clustering analysis revealed that IL-2M specifically expands multiple sub-states of Tregs with distinct expression profiles. TCR profiling with single-cell analysis uncovered Treg migration across tissues and transcriptional changes between clonally related Tregs after IL-2M treatment. Finally, we identified IL-2M–expanded Tnfrsf9+Il1rl1+ Tregs with superior suppressive function, highlighting the potential of IL-2M to expand highly suppressive Foxp3+ Tregs.
Collapse
Affiliation(s)
- Daniel R Lu
- Genome Analysis Unit, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Hao Wu
- Department of Oncology and Inflammation, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Ian Driver
- Genome Analysis Unit, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Sarah Ingersoll
- Department of Oncology and Inflammation, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Sue Sohn
- Department of Oncology and Inflammation, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Songli Wang
- Genome Analysis Unit, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Chi-Ming Li
- Genome Analysis Unit, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Hyewon Phee
- Department of Oncology and Inflammation, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| |
Collapse
|
23
|
Turpaev KT. Transcription Factor KLF2 and Its Role in the Regulation of Inflammatory Processes. BIOCHEMISTRY (MOSCOW) 2020; 85:54-67. [PMID: 32079517 DOI: 10.1134/s0006297920010058] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
KLF2 is a member of the Krüppel-like transcription factor family of proteins containing highly conserved DNA-binding zinc finger domains. KLF2 participates in the differentiation and regulation of the functional activity of monocytes, T lymphocytes, adipocytes, and vascular endothelial cells. The activity of KLF2 is controlled by several regulatory systems, including the MEKK2,3/MEK5/ERK5/MEF2 MAP kinase cascade, Rho family G-proteins, histone acetyltransferases CBP and p300, and histone deacetylases HDAC4 and HDAC5. Activation of KLF2 in endothelial cells induces eNOS expression and provides vasodilatory effect. Many KLF2-dependent genes participate in the suppression of blood coagulation and aggregation of T cells and macrophages with the vascular endothelium, thereby preventing atherosclerosis progression. KLF2 can have a dual effect on the gene transcription. Thus, it induces expression of multiple genes, but suppresses transcription of NF-κB-dependent genes. Transcription factors KLF2 and NF-κB are reciprocal antagonists. KLF2 inhibits induction of NF-κB-dependent genes, whereas NF-κB downregulates KLF2 expression. KLF2-mediated inhibition of NF-κB signaling leads to the suppression of cell response to the pro-inflammatory cytokines IL-1β and TNFα and results in the attenuation of inflammatory processes.
Collapse
Affiliation(s)
- K T Turpaev
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
24
|
Hyvärinen K, Koskela S, Niittyvuopio R, Nihtinen A, Volin L, Salmenniemi U, Putkonen M, Buño I, Gallardo D, Itälä-Remes M, Partanen J, Ritari J. Meta-Analysis of Genome-Wide Association and Gene Expression Studies Implicates Donor T Cell Function and Cytokine Pathways in Acute GvHD. Front Immunol 2020; 11:19. [PMID: 32117222 PMCID: PMC7008714 DOI: 10.3389/fimmu.2020.00019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022] Open
Abstract
Graft-vs.-host disease (GvHD) is a major complication after allogeneic hematopoietic stem cell transplantation that causes mortality and severe morbidity. Genetic disparities in human leukocyte antigens between the recipient and donor are known contributors to the risk of the disease. However, the overall impact of genetic component is complex, and consistent findings across different populations and studies remain sparse. To gain a comprehensive understanding of the genes responsible for GvHD, we combined genome-wide association studies (GWAS) from two distinct populations with previously published gene expression studies on GvHD in a single gene-level meta-analysis. We hypothesized that genes driving GvHD should be associated in both data modalities and therefore could be detected more readily through their combined effects in the integrated analysis rather than in separate analyses. The meta-analysis yielded a total of 51 acute GvHD-associated genes (false detection rate [FDR] <0.1). In support of our hypothesis, this number was significantly higher than that in a permutation meta-analysis involving the whole data set, as well as in separate meta-analyses on the GWAS and gene expression data sets. The genes indicated by the meta-analysis were significantly enriched in 277 Gene Ontology terms (FDR < 0.05), such as T cell function and cytokine-mediated signaling pathways, and the results highlighted several established immune mediators, such as interleukins and JAK-STAT signaling, and presented TRAF6 and TERT as potential effector candidates. Altogether, the results support the chosen methodological approach, implicate a role of gene-level variation in donors' key immunological regulators predisposing patients to acute GVHD, and present potential targets for therapeutic intervention.
Collapse
Affiliation(s)
| | - Satu Koskela
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - Riitta Niittyvuopio
- Stem Cell Transplantation Unit, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Anne Nihtinen
- Stem Cell Transplantation Unit, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Liisa Volin
- Stem Cell Transplantation Unit, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | | | | | - Ismael Buño
- Department of Hematology, Genomics Unit, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - David Gallardo
- Department of Hematology, Institut Català d'Oncologia, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
| | | | | | - Jarmo Ritari
- Finnish Red Cross Blood Service, Helsinki, Finland
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW To evaluate role of the lymph node in immune regulation and tolerance in transplantation and recent advances in the delivery of antigen and immune modulatory signals to the lymph node. RECENT FINDINGS Lymph nodes are a primary site of immune cell priming, activation, and modulation, and changes within the lymph node microenvironment have the potential to induce specific regulation, suppression, and potentially tolerance. Antigen enters the lymph node either from tissues via lymphatics, from blood via high endothelial venules, or directly via injection. Here we review different techniques and materials to deliver antigen to the lymph node including microparticles or nanoparticles, ex-vivo antigen presenting cell manipulation, and use of receptor conjugation for specific intralymph node targeting locations. SUMMARY The promising results point to powerful techniques to harness the lymph node microenvironment and direct systemic immune regulation. The materials, techniques, and approaches suggest that translational and clinical trials in nonhuman primate and patients may soon be possible.
Collapse
|
26
|
Rodríguez-Perea AL, Rojas M, Velilla-Hernández PA. High concentrations of atorvastatin reduce in-vitro function of conventional T and regulatory T cells. Clin Exp Immunol 2019; 196:237-248. [PMID: 30638266 DOI: 10.1111/cei.13260] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2019] [Indexed: 12/15/2022] Open
Abstract
Regulatory T cells (Tregs ) modulate the magnitude of immune responses and possess therapeutic potential in an array of immune diseases. Statins reduce the activation and proliferation of conventional T cells (Tcons ), and they seem to up-regulate the frequency and function of Tregs . However, there is a lack of simultaneous evaluation of the in-vitro effect of statins on the functional profile of Tregs versus Tcons . Herein, magnetically purified Tcons and Tregs were stimulated with CD3/CD28/interleukin (IL)-2 in the presence of atorvastatin (ATV) at 1 or 10 µM. The suppressive function of Tregs , the expression of markers associated with Treg function, activation levels, cytokine production and calcium flux in both subpopulations were assessed by flow cytometry. ATV had no cytotoxic effect on T cells at the concentrations used. Interestingly, 10 µM ATV hampered the suppressive capacity of Tregs . Moreover, this higher concentration reduced the expression of forkhead box protein 3 (FoxP3), cytotoxic T lymphocyte antigen (CTLA-4) and programmed death 1 (PD-1). In Tcons , ATV at 10 µM decreased PD-1 and CD45RO expression. The expression of CD25, CD69, CD95, CD38, CD62L, CCR7 and perforin was not affected in both subpopulations or at any ATV concentrations. Remarkably, 10 µM ATV increased the percentage of tumour necrosis factor (TNF)-α-producing Tregs . Although there was a reduction of calcium flux in Tcons and Tregs , it was only significant in 10 µM ATV-treated Tcons . These results suggested that 10 µM ATV affects the cellular functions of both populations; however, this concentration particularly affected several aspects of Treg biology: its suppressive function, cytokine production and expression of Treg -specific markers.
Collapse
Affiliation(s)
- A L Rodríguez-Perea
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia.,Grupo Bacterias & Cáncer, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - M Rojas
- Grupo de Inmunología Celular e Inmunogenética (GICIG), Facultad de Medicina, Instituto de Investigaciones Médicas, Universidad de Antioquia UdeA, Medellín, Colombia.,Unidad de Citometría, Facultad de Medicina, Sede de Investigación Universitaria, Universidad de Antioquia UdeA, Medellín, Colombia
| | - P A Velilla-Hernández
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
27
|
Overexpression of KLF14 protects against immune-mediated hepatic injury in mice. J Transl Med 2019; 99:37-47. [PMID: 30254317 DOI: 10.1038/s41374-018-0134-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022] Open
Abstract
The underlying immunopathogenic mechanisms of autoimmune hepatitis (AIH) have not yet been well elucidated. An impairment in regulatory T cells (Tregs) is key to the development of AIH. Krüppel-like factors (KLFs) regulate a broad of cellular processes including immunocyte maturation. KLF14 may regulate Treg differentiation, but the biological functions remain far from elucidated. In this study, we identified the hepatic expression of KLF14 in human and murine liver diseases. Immune-mediated hepatitis was induced by concanavalin A (Con A). A KLF14 recombinant adenoviruses plasmid (Ad-KLF14) was constructed and injected into mice. Tregs were assessed by flow cytometry analysis; inflammatory cytokines, such as tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6), were tested by enzyme-linked immunosorbent assay (ELISA). The expression of KLF14 was suppressed in a time-and dose-dependent manner. Changes in cytokine levels were consistent with the degree of hepatic injury. Overexpression of KLF14 protected the liver from immune-mediated damage in vivo. Ad-KLF14 transfection before Con A challenge increased the frequency of Tregs in liver mononuclear cells (MNCs), and suppressed the expression of cytokines. All of these improvements were completely abrogated after Treg deletion in vivo by intraperitoneal injection of a CD25 antibody. In conclusion, these data suggest that KLF14 plays an as-yet unrecognized role in immune-mediated hepatitis mainly via induced Treg differentiation. Our findings extend the knowledge of the biological function of KLF14 to the autoimmune disease field, and indicate the possibility of KLF14 as a therapeutic target in AIH patients.
Collapse
|
28
|
Hoeppli RE, MacDonald KN, Leclair P, Fung VCW, Mojibian M, Gillies J, Rahavi SMR, Campbell AIM, Gandhi SK, Pesenacker AM, Reid G, Lim CJ, Levings MK. Tailoring the homing capacity of human Tregs for directed migration to sites of Th1-inflammation or intestinal regions. Am J Transplant 2019; 19:62-76. [PMID: 29766641 DOI: 10.1111/ajt.14936] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 04/17/2018] [Accepted: 05/06/2018] [Indexed: 01/25/2023]
Abstract
Cell-based therapy with CD4+ FOXP3+ regulatory T cells (Tregs) is a promising strategy to limit organ rejection and graft-vs-host disease. Ongoing clinical applications have yet to consider how human Tregs could be modified to direct their migration to specific inflammation sites and/or tissues for more targeted immunosuppression. We show here that stable, homing-receptor-tailored human Tregs can be generated from thymic Tregs isolated from pediatric thymus or adult blood. To direct migration to Th1-inflammatory sites, addition of interferon-γ and IL-12 during Treg expansion produced suppressive, epigenetically stable CXCR3+ TBET+ FOXP3+ T helper (Th)1-Tregs. CXCR3 remained expressed after injection in vivo and Th1-Tregs migrated efficiently towards CXCL10 in vitro. To induce tissue-specific migration, addition of retinoic acid (RA) during Treg expansion induced expression of the gut-homing receptors α4β7-integrin and CCR9. FOXP3+ RA-Tregs had elevated expression of the functional markers latency-associated peptide and glycoprotein A repetitions predominant, increased suppressive capacity in vitro and migrated efficiently to healthy and inflamed intestine after injection into mice. Homing-receptor-tailored Tregs were epigenetically stable even after long-term exposure to inflammatory conditions, suppressive in vivo and characterized by Th1- or gut-homing-specific transcriptomes. Tailoring human thymic Treg homing during in vitro expansion offers a new and clinically applicable approach to improving the potency and specificity of Treg therapy.
Collapse
Affiliation(s)
- R E Hoeppli
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - K N MacDonald
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - P Leclair
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - V C W Fung
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - M Mojibian
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - J Gillies
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - S M R Rahavi
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - A I M Campbell
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - S K Gandhi
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - A M Pesenacker
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - G Reid
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - C J Lim
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - M K Levings
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
29
|
Mohamud R, LeMasurier JS, Boer JC, Sieow JL, Rolland JM, O'Hehir RE, Hardy CL, Plebanski M. Synthetic Nanoparticles That Promote Tumor Necrosis Factor Receptor 2 Expressing Regulatory T Cells in the Lung and Resistance to Allergic Airways Inflammation. Front Immunol 2017; 8:1812. [PMID: 29312323 PMCID: PMC5744007 DOI: 10.3389/fimmu.2017.01812] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/01/2017] [Indexed: 12/28/2022] Open
Abstract
Synthetic glycine coated 50 nm polystyrene nanoparticles (NP) (PS50G), unlike ambient NP, do not promote pulmonary inflammation, but instead, render lungs resistant to the development of allergic airway inflammation. In this study, we show that PS50G modulate the frequency and phenotype of regulatory T cells (Treg) in the lung, specifically increasing the proportion of tumor necrosis factor 2 (TNFR2) expressing Treg. Mice pre-exposed to PS50G, which were sensitized and then challenged with an allergen a month later, preferentially expanded TNFR2+Foxp3+ Treg, which further expressed enhanced levels of latency associated peptide and cytotoxic T-lymphocyte associated molecule-4. Moreover, PS50G-induced CD103+ dendritic cell activation in the lung was associated with the proliferative expansion of TNFR2+Foxp3+ Treg. These findings provide the first evidence that engineered NP can promote the selective expansion of maximally suppressing TNFR2+Foxp3+ Treg and further suggest a novel mechanism by which NP may promote healthy lung homeostasis.
Collapse
Affiliation(s)
- Rohimah Mohamud
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,CRC for Asthma and Airways, Sydney, NSW, Australia.,Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Jeanne S LeMasurier
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,CRC for Asthma and Airways, Sydney, NSW, Australia
| | - Jennifer C Boer
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Je Lin Sieow
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Jennifer M Rolland
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,CRC for Asthma and Airways, Sydney, NSW, Australia.,Department of Allergy, Immunology and Respiratory Medicine, Monash University and The Alfred Hospital, Melbourne, VIC, Australia
| | - Robyn E O'Hehir
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,CRC for Asthma and Airways, Sydney, NSW, Australia.,Department of Allergy, Immunology and Respiratory Medicine, Monash University and The Alfred Hospital, Melbourne, VIC, Australia
| | - Charles L Hardy
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,CRC for Asthma and Airways, Sydney, NSW, Australia.,Department of Allergy, Immunology and Respiratory Medicine, Monash University and The Alfred Hospital, Melbourne, VIC, Australia
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,School of Health and Biomedical Sciences, RMIT, Melbourne, VIC, Australia
| |
Collapse
|
30
|
Harnessing Advances in T Regulatory Cell Biology for Cellular Therapy in Transplantation. Transplantation 2017; 101:2277-2287. [PMID: 28376037 DOI: 10.1097/tp.0000000000001757] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cellular therapy with CD4FOXP3 T regulatory (Treg) cells is a promising strategy to induce tolerance after solid-organ transplantation or prevent graft-versus-host disease after transfer of hematopoietic stem cells. Treg cells currently used in clinical trials are either polyclonal, donor- or antigen-specific. Aside from variations in isolation and expansion protocols, however, most therapeutic Treg cell-based products are much alike. Ongoing basic science work has provided considerable new insight into multiple facets of Treg cell biology, including their stability, homing, and functional specialization; integrating these basic science discoveries with clinical efforts will support the development of next-generation therapeutic Treg cells with enhanced efficacy. In this review, we summarize recent advances in knowledge of how Treg cells home to lymphoid and peripheral tissues, and control antibody production and tissue repair. We also discuss newly appreciated pathways that modulate context-specific Treg cell function and stability. Strategies to improve and tailor Treg cells for cell therapy to induce transplantation tolerance are highlighted.
Collapse
|
31
|
Bin Dhuban K, d’Hennezel E, Nagai Y, Xiao Y, Shao S, Istomine R, Alvarez F, Ben-Shoshan M, Ochs H, Mazer B, Li B, Sekine C, Berezov A, Hancock W, Torgerson TR, Greene MI, Piccirillo CA. Suppression by human FOXP3
+
regulatory T cells requires FOXP3-TIP60 interactions. Sci Immunol 2017; 2. [DOI: 10.1126/sciimmunol.aai9297] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Targeting the FOXP3-TIP60 interaction may modulate T
reg
activity in IPEX and other autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Khalid Bin Dhuban
- Department of Microbiology and Immunology, McGill University and Research Institute of McGill University Health Centre, Montréal, Québec H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
| | - Eva d’Hennezel
- Department of Microbiology and Immunology, McGill University and Research Institute of McGill University Health Centre, Montréal, Québec H3A 2B4, Canada
| | - Yasuhiro Nagai
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104–6082, USA
| | - Yan Xiao
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104–6082, USA
| | - Steven Shao
- Department of Microbiology and Immunology, McGill University and Research Institute of McGill University Health Centre, Montréal, Québec H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
| | - Roman Istomine
- Department of Microbiology and Immunology, McGill University and Research Institute of McGill University Health Centre, Montréal, Québec H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University and Research Institute of McGill University Health Centre, Montréal, Québec H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
| | - Moshe Ben-Shoshan
- Division of Pediatric Allergy and Clinical Immunology, Department of Pediatrics, McGill University Health Center, Montréal, Québec H3H 1P3, Canada
| | - Hans Ochs
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98101–1304, USA
| | - Bruce Mazer
- Division of Pediatric Allergy and Clinical Immunology, Department of Pediatrics, McGill University Health Center, Montréal, Québec H3H 1P3, Canada
- FOCiS Centre of Excellence in Translational Immunology (CETI), Montréal, Québec H4A 3J1, Canada
| | - Bin Li
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104–6082, USA
| | | | - Alan Berezov
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104–6082, USA
| | - Wayne Hancock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104–6082, USA
| | - Troy R. Torgerson
- Division of Pediatric Allergy and Clinical Immunology, Department of Pediatrics, McGill University Health Center, Montréal, Québec H3H 1P3, Canada
| | - Mark I. Greene
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104–6082, USA
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University and Research Institute of McGill University Health Centre, Montréal, Québec H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
- FOCiS Centre of Excellence in Translational Immunology (CETI), Montréal, Québec H4A 3J1, Canada
- Division of Allergy and Clinical Immunology, Department of Medicine, McGill University and McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
32
|
Gregor CE, Foeng J, Comerford I, McColl SR. Chemokine-Driven CD4 + T Cell Homing: New Concepts and Recent Advances. Adv Immunol 2017; 135:119-181. [DOI: 10.1016/bs.ai.2017.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
33
|
Blanquiceth Y, Rodríguez-Perea AL, Tabares Guevara JH, Correa LA, Sánchez MD, Ramírez-Pineda JR, Velilla PA. Increase of Frequency and Modulation of Phenotype of Regulatory T Cells by Atorvastatin Is Associated with Decreased Lung Inflammatory Cell Infiltration in a Murine Model of Acute Allergic Asthma. Front Immunol 2016; 7:620. [PMID: 28066430 PMCID: PMC5174085 DOI: 10.3389/fimmu.2016.00620] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/06/2016] [Indexed: 01/24/2023] Open
Abstract
Regulatory T cells (Tregs) play an important role by controlling allergic inflammation of airways. Recently, it has been shown that statins have immunomodulatory properties, probably mediated by their effects on Tregs. Therefore, we evaluated the in vivo effect of atorvastatin (ATV) on Tregs and its association with the inflammatory process in a model of allergic asthma. BALB/c mice were sensitized with ovalbumin (OVA) and then challenged with intranasal OVA. ATV (40 mg/kg) was delivered by daily intraperitoneal injection for 7 or 15 days before each OVA challenge. ATV treatment for 7 days increased the frequency of Tregs in mediastinal lymph nodes (MLN) and the interleukin (IL)-10 in lungs. After 15 days of treatment, ATV increased the percentage of glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR+) and programmed cell death protein 1 (PD-1+) Tregs in the lung, without enhancing their suppressive activity, but also increased the percentage of conventional T cells expressing GITR+, PD1+, and OX-40 (tumor necrosis factor receptor superfamily member 4). Although no significant changes were observed in the number of inflammatory cells in the bronchoalveolar lavage (BAL), OVA-specific immunoglobulin E in the serum, and type 2 helper (Th2) cytokines in the lungs, there was a significant decrease of peribronchial inflammation that negatively correlated with the Tregs in MLN and the concentration of IL-10 in the lung. These results suggest that ATV has an immunomodulatory role possibly mediated by their effects on Tregs, which could contribute to the control of inflammation during allergic asthma. Further studies are necessary to elucidate the contribution of Treg to immunomodulatory action of statins in the context of allergic asthma.
Collapse
Affiliation(s)
- Yurany Blanquiceth
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA , Medellín , Colombia
| | - Ana Lucia Rodríguez-Perea
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA , Medellín , Colombia
| | - Jorge H Tabares Guevara
- Grupo Inmunomodulación, Facultad de Medicina, Universidad de Antioquia UdeA , Medellín , Colombia
| | - Luis Alfonso Correa
- Sección de Dermatología, Departamento de Medicina, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia; Laboratorio de Patología, Laboratorio Clínico VID, Obra de la Congregación Mariana, Medellín, Colombia
| | - María Dulfary Sánchez
- Stanley S. Scott Cancer Center & Louisiana Cancer Research Center, Health Sciences Center, Louisiana State University , New Orleans, LA , USA
| | | | - Paula Andrea Velilla
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA , Medellín , Colombia
| |
Collapse
|