1
|
Gong Y, Fei P, Zhang Y, Xu Y, Wei J. From Multi-Omics to Visualization and Beyond: Bridging Micro and Macro Insights in CAR-T Cell Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2501095. [PMID: 40349154 PMCID: PMC12120725 DOI: 10.1002/advs.202501095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/03/2025] [Indexed: 05/14/2025]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapies, a cornerstone of immunotherapy, have demonstrated remarkable efficacy in treating hematological malignancies and have more recently expanded into applications for solid tumors and autoimmune diseases. Emerging multidimensional profiling technologies offer promising solutions for enhancing CAR-T efficacy, overcoming resistance, and facilitating the development of novel CAR-T constructs. The integration of genomics, epigenomics, transcriptomics, proteomics, metabolomics, and microbiomics enables a comprehensive understanding of the intrinsic mechanisms underlying CAR-T therapy, while single-cell and spatial omics significantly improve data resolution and analytical depth. Coupled with advances in biomedical engineering, visualization technologies form the foundation for omics data generation by bridging microscopic and macroscopic scales and enabling dynamic, 3D in vivo monitoring of CAR-T behavior. Artificial intelligence (AI) further supports this framework by enabling the analysis of complex, high-dimensional datasets. This review highlights recent advances in the integration of multidimensional omics within CAR-T therapy and explores cutting-edge developments in visualization technologies and AI applications. The full convergence of multi-omics, visualization tools, and AI is poised to deliver transformative insights into the mechanisms governing CAR-T cell therapy.
Collapse
Affiliation(s)
- Yuting Gong
- Department of HematologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
- Immunotherapy Research Center for Hematologic Diseases of Hubei ProvinceWuhanHubei430030China
| | - Peng Fei
- Department of HematologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
- School of Optical and Electronic Information‐Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhanHubei430074China
- Advanced Biomedical Imaging FacilityHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Yicheng Zhang
- Department of HematologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
- Immunotherapy Research Center for Hematologic Diseases of Hubei ProvinceWuhanHubei430030China
- Key Laboratory of Organ TransplantationMinistry of EducationNHC Key Laboratory of Organ TransplantationKey Laboratory of Organ TransplantationChinese Academy of Medical SciencesWuhanHubei430030China
| | - Yang Xu
- National Clinical Research Center for Hematologic DiseasesJiangsu Institute of HematologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Institute of Blood and Marrow TransplantationSoochow UniversitySuzhouJiangsu215006China
| | - Jia Wei
- Department of HematologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430030China
- Immunotherapy Research Center for Hematologic Diseases of Hubei ProvinceWuhanHubei430030China
- Key Laboratory of Organ TransplantationMinistry of EducationNHC Key Laboratory of Organ TransplantationKey Laboratory of Organ TransplantationChinese Academy of Medical SciencesWuhanHubei430030China
| |
Collapse
|
2
|
Schmidleithner L, Stüve P, Feuerer M. Transposable elements as instructors of the immune system. Nat Rev Immunol 2025:10.1038/s41577-025-01172-3. [PMID: 40301669 DOI: 10.1038/s41577-025-01172-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2025] [Indexed: 05/01/2025]
Abstract
Transposable elements (TEs) are mobile repetitive nucleic acid sequences that have been incorporated into the genome through spontaneous integration, accounting for almost 50% of human DNA. Even though most TEs are no longer mobile today, studies have demonstrated that they have important roles in different biological processes, such as ageing, embryonic development, and cancer. TEs influence these processes through various mechanisms, including active transposition of TEs contributing to ongoing evolution, transposon transcription generating RNA or protein, and by influencing gene regulation as enhancers. However, how TEs interact with the immune system remains a largely unexplored field. In this Perspective, we describe how TEs might influence different aspects of the immune system, such as innate immune responses, T cell activation and differentiation, and tissue adaptation. Furthermore, TEs can serve as a source of neoantigens for T cells in antitumour immunity. We suggest that TE biology is an important emerging field of immunology and discuss the potential to harness the TE network therapeutically, for example, to improve immunotherapies for cancer and autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
| | - Philipp Stüve
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Markus Feuerer
- Leibniz Institute for Immunotherapy, Regensburg, Germany.
- Chair for Immunology, University Regensburg, Regensburg, Germany.
| |
Collapse
|
3
|
Norton EG, Chapman NM, Shi H, Meng X, Huang H, KC A, Rankin S, Saravia J, Yuan S, Hu H, Vogel P, Chi H. Vps34-orchestrated lipid signaling processes regulate the transitional heterogeneity and functional adaptation of effector regulatory T cells. PLoS Biol 2025; 23:e3003074. [PMID: 40215232 PMCID: PMC11990774 DOI: 10.1371/journal.pbio.3003074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/19/2025] [Indexed: 04/14/2025] Open
Abstract
Regulatory T cell (Treg) heterogeneity exists in lymphoid and non-lymphoid tissues, but we have limited understanding of context-dependent functions and spatiotemporal regulators of heterogenous Treg states, especially during perinatal life when immune tolerance is established. Here, we revealed that the class III PI3K Vps34 orchestrates effector Treg (eTreg) transitional heterogeneity during perinatal life. We found that loss of Vps34 reduced terminal eTreg accumulation in lymphoid tissues, associated with decreased Treg generation in non-lymphoid tissues and development of an early-onset autoimmune-like disease. After perinatal life, Vps34-deficient eTreg accumulation was further impaired due to reduced cell survival, highlighting temporal regulation of eTreg heterogeneity and maintenance by Vps34. Accordingly, inhibition of Vps34 in mature Tregs disrupted immune homeostasis but boosted anti-tumor immunity. Mechanistically, multiomics profiling approaches uncovered that Vps34-orchestrated transcriptional and epigenetic remodeling promotes terminal eTreg programming. Further, via genetic deletion of the Vps34-interacting proteins Atg14 or Uvrag in Tregs, we established that Atg14 but not Uvrag was required for the overall survival, but not terminal differentiation, of eTregs, suggesting that autophagy but not endocytosis partly contributed to Vps34-dependent effects. Accordingly, mice with Treg-specific loss of Atg14, but not Uvrag, had moderately disrupted immune homeostasis and reduced tumor growth, with Vps34- or Atg14-dependent gene signatures also being elevated in intratumoral Tregs from human cancer patients. Collectively, our study reveals distinct Vps34-orchestrated signaling events that regulate eTreg heterogeneity and functional adaptation and the pathophysiological consequences on autoimmunity versus anti-tumor immunity.
Collapse
Affiliation(s)
- Erienne G. Norton
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Nicole M. Chapman
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hao Shi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Xiaoxi Meng
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hongling Huang
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Anil KC
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Sherri Rankin
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Jordy Saravia
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Sujing Yuan
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Haoran Hu
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Peter Vogel
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hongbo Chi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
4
|
Tani-Ichi S, Abe S, Miyachi H, Kitano S, Shimba A, Ejima A, Hara T, Cui G, Kado T, Hori S, Tobe K, Ikuta K. IL-7Rα signaling in regulatory T cells of adipose tissue is essential for systemic glucose homeostasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:666-679. [PMID: 40107286 DOI: 10.1093/jimmun/vkae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/17/2024] [Indexed: 03/22/2025]
Abstract
Regulatory T cells (Tregs) mediate tissue homeostasis and repair. The function of the interleukin-7 receptor α (IL-7Rα) in nonlymphoid tissue Tregs is still unknown, although low expression of IL-7Rα is a widely accepted marker for Tregs. Here, we show that IL-33R (ST2)-expressing Tregs in the visceral adipose tissue (VAT) express the IL-7Rα at high levels. Treg-specific IL-7Rα-deficient mice exhibited reduced adipose ST2+ Tregs and impaired glucose tolerance, whereas IL-7Rα was dispensable for Tregs in lymphoid tissues. Mice deficient in thymic stromal lymphopoietin (TSLP), an additional ligand for IL-7Rα, displayed a modest decrease in adipose ST2+ Tregs and a reduced accumulation of adipose eosinophils, accompanied by slightly impaired glucose tolerance. In the VAT, mesothelial cells expressed IL-7, whereas adipose stem cells and folate receptor β-expressing tissue-resident macrophages expressed TSLP. Thus, this study indicates the significance of IL-7Rα signaling in the maintenance of VAT Tregs and glucose homeostasis, revealing a novel role for IL-7 and TSLP in immunometabolism.
Collapse
Affiliation(s)
- Shizue Tani-Ichi
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Abe
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hitoshi Miyachi
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Satsuki Kitano
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Akihiro Shimba
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aki Ejima
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Takahiro Hara
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Guangwei Cui
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tomonobu Kado
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Shohei Hori
- Laboratory of Immunology and Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki Tobe
- Research Center for Pre-Disease Science, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
5
|
Wang K, Farrell A, Zhou E, Qin H, Zeng Z, Zhou K, Cunha E Rocha K, Zhang D, Wang G, Atakilit A, Sheppard D, Lu LF, Jin C, Ying W. ATF4 drives regulatory T cell functional specification in homeostasis and obesity. Sci Immunol 2025; 10:eadp7193. [PMID: 40085690 DOI: 10.1126/sciimmunol.adp7193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Regulatory T cells (Tregs) have diverse functional specification in homeostasis and disease. However, how liver Tregs function and are transcriptionally regulated in obesity is not well understood. Here, we identified that effector Tregs expressing activating transcription factor 4 (ATF4) were enriched in the livers of obese mice. ATF4 was critical for driving an effector Treg transcriptional program, and ATF4-expressing Tregs promoted the development of obesity-induced liver fibrosis by enhancing transforming growth factor-β activation via integrin αvβ8. Treg-specific deletion of Atf4 resulted in reduced liver Tregs and attenuation of obesity-induced liver abnormalities. Furthermore, ATF4 was required to promote the differentiation of nonlymphoid tissue Treg precursors under steady state. These findings demonstrate that ATF4 is important for regulating Treg functional specification in homeostasis and obesity.
Collapse
Affiliation(s)
- Ke Wang
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Andrea Farrell
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Enchen Zhou
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Houji Qin
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Zixuan Zeng
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Kailun Zhou
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Karina Cunha E Rocha
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Dinghong Zhang
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Gaowei Wang
- Institute of Modern Biology, Nanjing University, Nanjing, China
| | - Amha Atakilit
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Dean Sheppard
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Li-Fan Lu
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Chunyu Jin
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
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
|
7
|
Marin-Rodero M, Cintado E, Walker AJ, Jayewickreme T, Pinho-Ribeiro FA, Richardson Q, Jackson R, Chiu IM, Benoist C, Stevens B, Trejo JL, Mathis D. The meninges host a distinct compartment of regulatory T cells that preserves brain homeostasis. Sci Immunol 2025; 10:eadu2910. [PMID: 39873623 PMCID: PMC11924117 DOI: 10.1126/sciimmunol.adu2910] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025]
Abstract
Our understanding of the meningeal immune system has recently burgeoned, particularly regarding how innate and adaptive effector cells are mobilized to meet brain challenges. However, information on how meningeal immunocytes guard brain homeostasis in healthy individuals remains limited. This study highlights the heterogeneous, polyfunctional regulatory T cell (Treg) compartment in the meninges. A Treg subtype specialized in controlling interferon-γ (IFN-γ) responses and another dedicated to regulating follicular B cell responses were substantial components of this compartment. Accordingly, punctual Treg ablation rapidly unleashed IFN-γ production by meningeal lymphocytes, unlocked access to the brain parenchyma, and altered meningeal B cell profiles. Distally, the hippocampus assumed a reactive state, with morphological and transcriptional changes in multiple glial cell types. Within the dentate gyrus, neural stem cells underwent more death and were blocked from further differentiation, which coincided with impairments in short-term spatial-reference memory. Thus, meningeal Tregs are a multifaceted safeguard of brain homeostasis at steady state.
Collapse
Affiliation(s)
| | - Elisa Cintado
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas; Madrid, Spain
| | - Alec J. Walker
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | | | | | | | - Ruaidhrí Jackson
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| | - Isaac M. Chiu
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| | | | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School; Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital; Boston, MA, USA
| | - José Luís Trejo
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas; Madrid, Spain
| | - Diane Mathis
- Department of Immunology, Harvard Medical School; Boston, MA, USA
| |
Collapse
|
8
|
Revilla SA, Frederiks CL, Prekovic S, Mocholi E, Kranenburg O, Coffer PJ. Tumor-derived colorectal cancer organoids induce a unique Treg cell population by directing CD4 + T cell differentiation. iScience 2025; 28:111827. [PMID: 39995881 PMCID: PMC11848486 DOI: 10.1016/j.isci.2025.111827] [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: 07/30/2024] [Revised: 11/22/2024] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
In colorectal cancer (CRC), increased numbers of tumor-infiltrating CD4+ regulatory T (Treg) cells correlate with tumor development, immunotherapy failure, and poor prognosis. To assess how CRC tumors directly modulate Treg cell differentiation, we developed an in vitro co-culture system using CD4+ T cells from Foxp3eGFP mice and CRC tumor-derived organoids. Co-culture resulted in a significant increase in Treg cell numbers. RNA-sequencing identified a distinct transcriptional profile of CRC organoid-induced Treg cells, with upregulation of genes associated with CRC Treg cells in vivo. High expression of genes upregulated in CRC organoid-induced Treg cells correlates with shorter progression-free intervals and overall survival in CRC patients. Human CRC organoids similarly induced Treg cells with enhanced suppressive capacity and upregulated genes linked to CRC Treg cells in vivo. This model provides insights into how CRC tumors modulate CD4+ T cell differentiation and can identify approaches to disrupt Treg cells and stimulate anti-tumor immunity.
Collapse
Affiliation(s)
- Sonia Aristin Revilla
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
- Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cynthia L. Frederiks
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
- Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Stefan Prekovic
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Enric Mocholi
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Onno Kranenburg
- Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Paul J. Coffer
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
9
|
Boccardo S, Rodriguez C, Gimenez CMS, Araujo Furlan CL, Abrate CP, Almada L, Saldivia Concepción MA, Skewes-Cox P, Rao SPS, Mukdsi JH, Montes CL, Gruppi A, Acosta Rodríguez EV. Dynamics of tissue repair regulatory T cells and damage in acute Trypanosoma cruzi infection. PLoS Pathog 2025; 21:e1012906. [PMID: 39883714 PMCID: PMC11813105 DOI: 10.1371/journal.ppat.1012906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 02/11/2025] [Accepted: 01/14/2025] [Indexed: 02/01/2025] Open
Abstract
Tissue-repair regulatory T cells (trTregs) comprise a specialized cell subset essential for tissue homeostasis and repair. While well-studied in sterile injury models, their role in infection-induced tissue damage and antimicrobial immunity is less understood. We investigated trTreg dynamics during acute Trypanosoma cruzi infection, marked by extensive tissue damage and strong CD8+ immunity. Unlike sterile injury models, trTregs significantly declined in secondary lymphoid organs and non-lymphoid target tissues during infection, correlating with systemic and local tissue damage, and downregulation of function-associated genes in skeletal muscle. This decline was linked to decreased systemic IL-33 levels, a key trTreg growth factor, and promoted by the Th1 cytokine IFN-γ. Early recombinant IL-33 treatment increased trTregs, type 2 innate lymphoid cells, and parasite-specific CD8+ cells at specific time points after infection, leading to reduced tissue damage, lower parasite burden, and improved disease outcome. Our findings not only provide novel insights into trTregs during infection but also highlight the potential of optimizing immune balance by modulating trTreg responses to promote tissue repair while maintaining effective pathogen control during infection-induced injury.
Collapse
Affiliation(s)
- Santiago Boccardo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | - Constanza Rodriguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | - Camila M. S. Gimenez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | - Cintia L. Araujo Furlan
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | - Carolina P. Abrate
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | - Laura Almada
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | | | - Peter Skewes-Cox
- BioMedical Research, Novartis, Emeryville, California, United States of America
| | - Srinivasa P. S. Rao
- BioMedical Research, Novartis, Emeryville, California, United States of America
| | - Jorge H. Mukdsi
- Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET). Córdoba, Argentina
- Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | - Carolina L. Montes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| | - Eva V. Acosta Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET). Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba. Córdoba, Argentina
| |
Collapse
|
10
|
Hattori K, Tanaka S, Hashiba D, Tamura J, Etori K, Kageyama T, Ito T, Meguro K, Iwata A, Suto A, Suzuki K, Nakamura J, Ohtori S, Ziegler SF, Nakajima H. Synovial regulatory T cells expressing ST2 deteriorate joint inflammation through the suppression of immunoregulatory eosinophils. J Autoimmun 2024; 149:103333. [PMID: 39509740 DOI: 10.1016/j.jaut.2024.103333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic polyarthritis. It is well-established that helper T cells play crucial roles in the development and deterioration of RA. Recent studies also revealed the significant roles of regulatory T (Treg) cells in this context. Although Treg cells distributed in peripheral tissues exhibit various functions, the characteristics of synovial Treg cells remain unknown. In this study, we demonstrate that synovial Treg cells exacerbate synovial inflammation by reducing the number of immunoregulatory eosinophils through competitive consumption of IL-33. Synovial Treg cells expressed ST2 in a murine arthritis model, and surprisingly, Treg-specific ST2 knockout (ST2ΔTreg) mice exhibited attenuated arthritis. In ST2ΔTreg mice, an increase in immunoregulatory synovial eosinophils was observed. Additionally, immunoregulatory eosinophils were found to express ST2, and ST2-expressing Treg cells controlled the abundance of immunoregulatory eosinophils, possibly by consuming IL-33. Our results highlight that a subset of synovial Treg cells possesses the machinery to worsen arthritis by suppressing eosinophils. In the future landscape where Treg cell-based therapies are employed for autoimmune diseases, it is important to comprehend the characteristics of disease-related Treg cells. Understanding these aspects is crucial for ensuring safer treatment modalities that do not inadvertently worsen the diseases.
Collapse
MESH Headings
- Animals
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Eosinophils/immunology
- Eosinophils/metabolism
- Mice
- Interleukin-1 Receptor-Like 1 Protein/metabolism
- Interleukin-1 Receptor-Like 1 Protein/genetics
- Mice, Knockout
- Interleukin-33/metabolism
- Interleukin-33/immunology
- Interleukin-33/genetics
- Synovial Membrane/immunology
- Synovial Membrane/pathology
- Synovial Membrane/metabolism
- Disease Models, Animal
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Humans
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Koto Hattori
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Daisuke Hashiba
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan; Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Jun Tamura
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Keishi Etori
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Kazuyuki Meguro
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Arifumi Iwata
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Akira Suto
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Kotaro Suzuki
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Junichi Nakamura
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Seiji Ohtori
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| | - Steven F Ziegler
- Center for Fundamental Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, 98101-2795, USA.
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba City, Chiba, 260-8670, Japan.
| |
Collapse
|
11
|
Costa CE, Watowich MM, Goldman EA, Sterner KN, Negron-Del Valle JE, Phillips D, Platt ML, Montague MJ, Brent LJN, Higham JP, Snyder-Mackler N, Lea AJ. Genetic Architecture of Immune Cell DNA Methylation in the Rhesus Macaque. Mol Ecol 2024:e17576. [PMID: 39582237 DOI: 10.1111/mec.17576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/23/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
Genetic variation that impacts gene regulation, rather than protein function, can have strong effects on trait variation both within and between species. Epigenetic mechanisms, such as DNA methylation, are often an important intermediate link between genotype and phenotype, yet genetic effects on DNA methylation remain understudied in natural populations. To address this gap, we used reduced representation bisulfite sequencing to measure DNA methylation levels at 555,856 CpGs in peripheral whole blood of 573 samples collected from free-ranging rhesus macaques (Macaca mulatta) living on the island of Cayo Santiago, Puerto Rico. We used allele-specific methods to map cis-methylation quantitative trait loci (meQTL) and tested for effects of 243,389 single nucleotide polymorphisms (SNPs) on local DNA methylation levels. Of 776,092 tested SNP-CpG pairs, we identified 516,213 meQTL, with 69.12% of CpGs having at least one meQTL (FDR < 5%). On average, meQTL explained 21.2% of nearby methylation variance, significantly more than age or sex. meQTL were enriched in genomic compartments where methylation is likely to impact gene expression, for example, promoters, enhancers and binding sites for methylation-sensitive transcription factors. In support, using mRNA-seq data from 172 samples, we confirmed 332 meQTL as whole blood cis-expression QTL (eQTL) in the population, and found meQTL-eQTL genes were enriched for immune response functions, like antigen presentation and inflammation. Overall, our study takes an important step towards understanding the genetic architecture of DNA methylation in natural populations, and more generally points to the biological mechanisms driving phenotypic variation in our close relatives.
Collapse
Affiliation(s)
- Christina E Costa
- Department of Anthropology, New York University, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Marina M Watowich
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Kirstin N Sterner
- Department of Anthropology, University of Oregon, Eugene, Oregon, USA
| | - Josue E Negron-Del Valle
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
| | - Daniel Phillips
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - James P Higham
- Department of Anthropology, New York University, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Noah Snyder-Mackler
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
- Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Amanda J Lea
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
12
|
Geiselhöringer AL, Kolland D, Patt AJ, Hammann L, Köhler A, Kreft L, Wichmann N, Hils M, Ruedl C, Riemann M, Biedermann T, Anz D, Diefenbach A, Voehringer D, Schmidt-Weber CB, Straub T, Pasztoi M, Ohnmacht C. Dominant immune tolerance in the intestinal tract imposed by RelB-dependent migratory dendritic cells regulates protective type 2 immunity. Nat Commun 2024; 15:9143. [PMID: 39443450 PMCID: PMC11500181 DOI: 10.1038/s41467-024-53112-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Dendritic cells (DCs) are crucial for initiating protective immune responses and have also been implicated in the generation and regulation of Foxp3+ regulatory T cells (Treg cells). Here, we show that in the lamina propria of the small intestine, the alternative NF-κB family member RelB is necessary for the differentiation of cryptopatch and isolated lymphoid follicle-associated DCs (CIA-DCs). Moreover, single-cell RNA sequencing reveals a RelB-dependent signature in migratory DCs in mesenteric lymph nodes favoring DC-Treg cell interaction including elevated expression and release of the chemokine CCL22 from RelB-deficient conventional DCs (cDCs). In line with the key role of CCL22 to facilitate DC-Treg cell interaction, RelB-deficient DCs have a selective advantage to interact with Treg cells in an antigen-specific manner. In addition, DC-specific RelB knockout animals show increased total Foxp3+ Treg cell numbers irrespective of inflammatory status. Consequently, DC-specific RelB knockout animals fail to mount protective Th2-dominated immune responses in the intestine after infection with Heligmosomoides polygyrus bakeri. Thus, RelB expression in cDCs acts as a rheostat to establish a tolerogenic set point that is maintained even during strong type 2 immune conditions and thereby is a key regulator of intestinal homeostasis.
Collapse
Affiliation(s)
- Anna-Lena Geiselhöringer
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Arisha Johanna Patt
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Linda Hammann
- Division of Clinical Pharmacology, LMU University Hospital, LMU, Munich, Germany
| | - Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Luisa Kreft
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076, Tuebingen, Germany
| | - Nina Wichmann
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Miriam Hils
- Department of Dermatology and Allergy Biederstein, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Marc Riemann
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745, Jena, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - David Anz
- Division of Clinical Pharmacology, LMU University Hospital, LMU, Munich, Germany
- Department of Medicine II, LMU University Hospital, LMU, Munich, Germany
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, 12203, Berlin, Germany
- Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, an Institute of the Leibniz Association, 10117, Berlin, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, 91054, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Tobias Straub
- Bioinformatics Core Unit, Biomedical Center, Ludwig-Maximilians-University, 82152, Planegg, Germany
| | - Maria Pasztoi
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany.
| |
Collapse
|
13
|
Byun S, Lee J, Choi YH, Ko H, Lee C, Park JC, Kim SW, Lee H, Sharma A, Kim KS, Rudra D, Kim JK, Im SH. Gut Microbiota Defines Functional Direction of Colonic Regulatory T Cells with Unique TCR Repertoires. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:886-897. [PMID: 39101764 DOI: 10.4049/jimmunol.2300395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/03/2024] [Indexed: 08/06/2024]
Abstract
Intestinal microbiota and selected strains of commensal bacteria influence regulatory T (Treg) cell functionality in the colon. Nevertheless, whether and how microbiota changes the transcriptome profile and TCR specificities of colonic Tregs remain to be precisely defined. In this study, we have employed single-cell RNA sequencing and comparatively analyzed colonic Tregs from specific pathogen-free and germ-free (GF) mice. We found that microbiota shifts the activation trajectory of colonic Tregs toward a distinct phenotypic subset enriched in specific pathogen-free but not in GF mice. Moreover, microbiota induced the expansion of specific Treg clonotypes with shared transcriptional specificities. The microbiota-induced subset of colonic Tregs, identified as PD-1- CXCR3+ Tregs, displayed enhanced suppressive capabilities compared with colonic Tregs derived from GF mice, enhanced production of IL-10, and were the primary regulators of enteric inflammation in dextran sodium sulfate-induced colitis. These findings identify a hitherto unknown gut microbiota and immune cell interaction module that could contribute to the development of a therapeutic modality for intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Seohyun Byun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jusung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Republic of Korea
| | - Yoon Ha Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Republic of Korea
| | - Haeun Ko
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Changhon Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - John Chulhoon Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Seung Won Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Haena Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Amit Sharma
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dipayan Rudra
- School of Life Science and Technology, ShanghaiTech University; Shanghai, People's Republic of China
| | - Jong Kyoung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
- ImmunoBiome Inc., Pohang, Republic of Korea
| |
Collapse
|
14
|
Lui PP, Xu JZ, Aziz H, Sen M, Ali N. Jagged-1+ skin Tregs modulate cutaneous wound healing. Sci Rep 2024; 14:20999. [PMID: 39251686 PMCID: PMC11385218 DOI: 10.1038/s41598-024-71512-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024] Open
Abstract
Skin-resident regulatory T cells (Tregs) play an irreplaceable role in orchestrating cutaneous immune homeostasis and repair, including the promotion of hair regeneration via the Notch signaling ligand Jagged-1 (Jag1). While skin Tregs are indispensable for facilitating tissue repair post-wounding, it remains unknown if Jag1-expressing skin Tregs impact wound healing. Using a tamoxifen inducible Foxp3creERT2Jag1fl/fl model, we show that loss of functional Jag1 in Tregs significantly delays the rate of full-thickness wound closure. Unlike in hair regeneration, skin Tregs do not utilize Jag1 to impact epithelial stem cells during wound healing. Instead, mice with Treg-specific Jag1 ablation exhibit a significant reduction in Ly6G + neutrophil accumulation at the wound site. However, during both homeostasis and wound healing, the loss of Jag1 in Tregs does not impact the overall abundance or activation profile of immune cell targets in the skin, such as CD4+ and CD8+ T cells, or pro-inflammatory macrophages. This collectively suggests that skin Tregs may utilize Jag1-Notch signalling to co-ordinate innate cell recruitment under conditions of injury but not homeostasis. Overall, our study demonstrates the importance of Jag1 expression in Tregs to facilitate adequate wound repair in the skin.
Collapse
Affiliation(s)
- Prudence PokWai Lui
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK
| | - Jessie Z Xu
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK
| | - Hafsah Aziz
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK
| | - Monica Sen
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK
| | - Niwa Ali
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK.
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK.
| |
Collapse
|
15
|
Okamoto M, Kuratani A, Okuzaki D, Kamiyama N, Kobayashi T, Sasai M, Yamamoto M. Tff1-expressing Tregs in lung prevent exacerbation of Bleomycin-induced pulmonary fibrosis. Front Immunol 2024; 15:1440918. [PMID: 39286257 PMCID: PMC11402662 DOI: 10.3389/fimmu.2024.1440918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Bleomycin (BLM) induces lung injury, leading to inflammation and pulmonary fibrosis. Regulatory T cells (Tregs) maintain self-tolerance and control host immune responses. However, little is known about their involvement in the pathology of pulmonary fibrosis. Here we show that a unique Treg subset expressing trefoil factor family 1 (Tff1) emerges in the BLM-injured lung. These Tff1-expressing Tregs (Tff1-Tregs) were induced by IL-33. Moreover, although Tff1 ablation in Tregs did not change the pathological condition, selective ablation of Tff1-Tregs using an intersectional genetic method promoted pro-inflammatory features of macrophages in the injured lung and exacerbated the fibrosis. Taken together, our study revealed the presence of a unique Treg subset expressing Tff1 in BLM-injured lungs and their critical role in the injured lung to ameliorate fibrosis.
Collapse
Affiliation(s)
- Masaaki Okamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Immunoparasitology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ayumi Kuratani
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Immunoparasitology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Osaka University, Suita, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
- Research Center for GLOBAL and LOCAL Infectious Diseases, Oita University, Oita, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Immunoparasitology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Immunoparasitology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Suita, Japan
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| |
Collapse
|
16
|
Schmidleithner L, Stüve P, Feuerer M. FOXP3 snatches transcription factors depending on the context. J Exp Med 2024; 221:e20240940. [PMID: 38949640 PMCID: PMC11215522 DOI: 10.1084/jem.20240940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
FOXP3 hijacks DNA-binding proteins to regulate gene expression. In this issue of JEM, He et al. (https://doi.org/10.1084/jem.20232068) propose a dynamic model in which FOXP3 associates with DNA-binding proteins to regulate Treg cell function in response to environmental cues.
Collapse
Affiliation(s)
- Lisa Schmidleithner
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Philipp Stüve
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Markus Feuerer
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| |
Collapse
|
17
|
Khan A, Roy P, Ley K. Breaking tolerance: the autoimmune aspect of atherosclerosis. Nat Rev Immunol 2024; 24:670-679. [PMID: 38472321 PMCID: PMC11682649 DOI: 10.1038/s41577-024-01010-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is a chronic inflammatory disease of the arterial walls and is characterized by the accumulation of lipoproteins that are insufficiently cleared by phagocytes. Following the initiation of atherosclerosis, the pathological progression is accelerated by engagement of the adaptive immune system. Atherosclerosis triggers the breakdown of tolerance to self-components. This loss of tolerance is reflected in defective expression of immune checkpoint molecules, dysfunctional antigen presentation, and aberrations in T cell populations - most notably in regulatory T (Treg) cells - and in the production of autoantibodies. The breakdown of tolerance to self-proteins that is observed in ASCVD may be linked to the conversion of Treg cells to 'exTreg' cells because many Treg cells in ASCVD express T cell receptors that are specific for self-epitopes. Alternatively, or in addition, breakdown of tolerance may trigger the activation of naive T cells, resulting in the clonal expansion of T cell populations with pro-inflammatory and cytotoxic effector phenotypes. In this Perspective, we review the evidence that atherosclerosis is associated with a breakdown of tolerance to self-antigens, discuss possible immunological mechanisms and identify knowledge gaps to map out future research. Rational approaches aimed at re-establishing immune tolerance may become game changers in treating ASCVD and in preventing its downstream sequelae, which include heart attacks and strokes.
Collapse
Affiliation(s)
- Amir Khan
- Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Payel Roy
- Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Klaus Ley
- Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
18
|
Simon M, Stüve P, Schmidleithner L, Bittner S, Beumer N, Strieder N, Schmidl C, Pant A, Gebhard C, Eigenberger A, Rehli M, Prantl L, Hehlgans T, Brors B, Imbusch CD, Delacher M, Feuerer M. Single-cell chromatin accessibility and transposable element landscapes reveal shared features of tissue-residing immune cells. Immunity 2024; 57:1975-1993.e10. [PMID: 39047731 DOI: 10.1016/j.immuni.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/12/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024]
Abstract
Tissue adaptation is required for regulatory T (Treg) cell function within organs. Whether this program shares aspects with other tissue-localized immune populations is unclear. Here, we analyzed single-cell chromatin accessibility data, including the transposable element (TE) landscape of CD45+ immune cells from colon, skin, adipose tissue, and spleen. We identified features of organ-specific tissue adaptation across different immune cells. Focusing on tissue Treg cells, we found conservation of the Treg tissue adaptation program in other tissue-localized immune cells, such as amphiregulin-producing T helper (Th)17 cells. Accessible TEs can act as regulatory elements, but their contribution to tissue adaptation is not understood. TE landscape analysis revealed an enrichment of specific transcription factor binding motifs in TE regions within accessible chromatin peaks. TEs, specifically from the LTR family, were located in enhancer regions and associated with tissue adaptation. These findings broaden our understanding of immune tissue residency and provide an important step toward organ-specific immune interventions.
Collapse
Affiliation(s)
- Malte Simon
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany; Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany
| | - Philipp Stüve
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany
| | - Lisa Schmidleithner
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany
| | - Sebastian Bittner
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany
| | - Niklas Beumer
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany; DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, 68167 Mannheim, Germany; Division of Personalized Medical Oncology, DKFZ, 69120 Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | | | | | - Asmita Pant
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany
| | - Claudia Gebhard
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany
| | - Andreas Eigenberger
- Department of Plastic, Hand, and Reconstructive Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Michael Rehli
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany; Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand, and Reconstructive Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Thomas Hehlgans
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, 69120 Heidelberg, Germany; Medical Faculty Heidelberg and Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Charles D Imbusch
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Michael Delacher
- Institute of Immunology, University Medical Center Mainz, 55131 Mainz, Germany; Research Center for Immunotherapy, University Medical Center Mainz, 55131 Mainz, Germany
| | - Markus Feuerer
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany; Chair for Immunology, University Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
19
|
Wells AC, Lima-Junior DS, Link VM, Smelkinson M, Krishnamurthy SR, Chi L, Segrist E, Rivera CA, Teijeiro A, Bouladoux N, Belkaid Y. Adaptive immunity to retroelements promotes barrier integrity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.606346. [PMID: 39149266 PMCID: PMC11326312 DOI: 10.1101/2024.08.09.606346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Maintenance of tissue integrity is a requirement of host survival. This mandate is of prime importance at barrier sites that are constitutively exposed to the environment. Here, we show that exposure of the skin to non-inflammatory xenobiotics promotes tissue repair; more specifically, mild detergent exposure promotes the reactivation of defined retroelements leading to the induction of retroelement-specific CD8+ T cells. These T cell responses are Langerhans cell dependent and establish tissue residency within the skin. Upon injury, retroelement-specific CD8+ T cells significantly accelerate wound repair via IL-17A. Collectively, this work demonstrates that tonic environmental exposures and associated adaptive responses to retroelements can be coopted to preemptively set the tissue for maximal resilience to injury.
Collapse
Affiliation(s)
- Alexandria C. Wells
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Djalma Souza Lima-Junior
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Verena M. Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Margery Smelkinson
- Biological Imaging, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Siddharth R. Krishnamurthy
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Liang Chi
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elisha Segrist
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Claudia A. Rivera
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ana Teijeiro
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
20
|
Burton OT, Bricard O, Tareen S, Gergelits V, Andrews S, Biggins L, Roca CP, Whyte C, Junius S, Brajic A, Pasciuto E, Ali M, Lemaitre P, Schlenner SM, Ishigame H, Brown BD, Dooley J, Liston A. The tissue-resident regulatory T cell pool is shaped by transient multi-tissue migration and a conserved residency program. Immunity 2024; 57:1586-1602.e10. [PMID: 38897202 DOI: 10.1016/j.immuni.2024.05.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/27/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The tissues are the site of many important immunological reactions, yet how the immune system is controlled at these sites remains opaque. Recent studies have identified Foxp3+ regulatory T (Treg) cells in non-lymphoid tissues with unique characteristics compared with lymphoid Treg cells. However, tissue Treg cells have not been considered holistically across tissues. Here, we performed a systematic analysis of the Treg cell population residing in non-lymphoid organs throughout the body, revealing shared phenotypes, transient residency, and common molecular dependencies. Tissue Treg cells from different non-lymphoid organs shared T cell receptor (TCR) sequences, with functional capacity to drive multi-tissue Treg cell entry and were tissue-agnostic on tissue homing. Together, these results demonstrate that the tissue-resident Treg cell pool in most non-lymphoid organs, other than the gut, is largely constituted by broadly self-reactive Treg cells, characterized by transient multi-tissue migration. This work suggests common regulatory mechanisms may allow pan-tissue Treg cells to safeguard homeostasis across the body.
Collapse
Affiliation(s)
- Oliver T Burton
- Department of Pathology, University of Cambridge, Cambridge, UK; VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium; Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Orian Bricard
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Samar Tareen
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Vaclav Gergelits
- Department of Pathology, University of Cambridge, Cambridge, UK; Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Simon Andrews
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Laura Biggins
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Carlos P Roca
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Carly Whyte
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Steffie Junius
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Aleksandra Brajic
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Emanuela Pasciuto
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium; University of Antwerp, Center of Molecular Neurology, Antwerp, Belgium
| | - Magda Ali
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Pierre Lemaitre
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Susan M Schlenner
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium
| | - Harumichi Ishigame
- Laboratory for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan; Near-InfraRed Photo-Immunotherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Brian D Brown
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James Dooley
- Department of Pathology, University of Cambridge, Cambridge, UK; VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium; Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Adrian Liston
- Department of Pathology, University of Cambridge, Cambridge, UK; VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven, University of Leuven, Department of Microbiology and Immunology, Leuven, Belgium; Babraham Institute, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
21
|
Pacella I, Pinzon Grimaldos A, Rossi A, Tucci G, Zagaglioni M, Potenza E, Pinna V, Rotella I, Cammarata I, Cancila V, Belmonte B, Tripodo C, Pietropaolo G, Di Censo C, Sciumè G, Licursi V, Peruzzi G, Antonucci Y, Campello S, Guerrieri F, Iebba V, Prota R, Di Chiara M, Terrin G, De Peppo V, Grazi GL, Barnaba V, Piconese S. Iron capture through CD71 drives perinatal and tumor-associated Treg expansion. JCI Insight 2024; 9:e167967. [PMID: 38954474 PMCID: PMC11383606 DOI: 10.1172/jci.insight.167967] [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: 12/12/2022] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Besides suppressing immune responses, regulatory T cells (Tregs) maintain tissue homeostasis and control systemic metabolism. Whether iron is involved in Treg-mediated tolerance is completely unknown. Here, we showed that the transferrin receptor CD71 was upregulated on activated Tregs infiltrating human liver cancer. Mice with a Treg-restricted CD71 deficiency spontaneously developed a scurfy-like disease, caused by impaired perinatal Treg expansion. CD71-null Tregs displayed decreased proliferation and tissue-Treg signature loss. In perinatal life, CD71 deficiency in Tregs triggered hepatic iron overload response, characterized by increased hepcidin transcription and iron accumulation in macrophages. Lower bacterial diversity, and reduction of beneficial species, were detected in the fecal microbiota of CD71 conditional knockout neonates. Our findings indicate that CD71-mediated iron absorption is required for Treg perinatal expansion and is related to systemic iron homeostasis and bacterial gut colonization. Therefore, we hypothesize that Tregs establish nutritional tolerance through competition for iron during bacterial colonization after birth.
Collapse
Affiliation(s)
- Ilenia Pacella
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Alessandra Rossi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Gloria Tucci
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marta Zagaglioni
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Elena Potenza
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Valeria Pinna
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ivano Rotella
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ilenia Cammarata
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | | | - Chiara Di Censo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Sciumè
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Valerio Licursi
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy, Sapienza University of Rome, Rome, Italy
| | - Giovanna Peruzzi
- Centre for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Ylenia Antonucci
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Silvia Campello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Guerrieri
- Cancer Research Centre of Lyon (CRCL), UMR Inserm U1052/CNRS 5286, Lyon, France
| | - Valerio Iebba
- Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy
| | - Rita Prota
- Department of Maternal and Child Health, Sapienza University of Rome, Rome, Italy
| | - Maria Di Chiara
- Department of Maternal and Child Health, Sapienza University of Rome, Rome, Italy
| | - Gianluca Terrin
- Department of Maternal and Child Health, Sapienza University of Rome, Rome, Italy
| | - Valerio De Peppo
- Hepatobiliary and Pancreatic Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gian Luca Grazi
- Hepatobiliary and Pancreatic Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Vincenzo Barnaba
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Piconese
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
- Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
22
|
Huang B, Bopp T, Li G. 8th Sino-German symposium on immunology: fostering mutual trust and collaborative endeavors for advancing immunological science. Cell Mol Immunol 2024; 21:798-799. [PMID: 38822078 PMCID: PMC11214616 DOI: 10.1038/s41423-024-01173-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/02/2024] Open
Affiliation(s)
- Bo Huang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Tobias Bopp
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany.
| | - Guideng Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.
- Key Laboratory of Synthetic Biology Regulatory Element, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.
| |
Collapse
|
23
|
Ramírez-Valle F, Maranville JC, Roy S, Plenge RM. Sequential immunotherapy: towards cures for autoimmunity. Nat Rev Drug Discov 2024; 23:501-524. [PMID: 38839912 DOI: 10.1038/s41573-024-00959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 06/07/2024]
Abstract
Despite major progress in the treatment of autoimmune diseases in the past two decades, most therapies do not cure disease and can be associated with increased risk of infection through broad suppression of the immune system. However, advances in understanding the causes of autoimmune disease and clinical data from novel therapeutic modalities such as chimeric antigen receptor T cell therapies provide evidence that it may be possible to re-establish immune homeostasis and, potentially, prolong remission or even cure autoimmune diseases. Here, we propose a 'sequential immunotherapy' framework for immune system modulation to help achieve this ambitious goal. This framework encompasses three steps: controlling inflammation; resetting the immune system through elimination of pathogenic immune memory cells; and promoting and maintaining immune homeostasis via immune regulatory agents and tissue repair. We discuss existing drugs and those in development for each of the three steps. We also highlight the importance of causal human biology in identifying and prioritizing novel immunotherapeutic strategies as well as informing their application in specific patient subsets, enabling precision medicine approaches that have the potential to transform clinical care.
Collapse
|
24
|
Marin-Rodero M, Reyes EC, Walker AJ, Jayewickreme T, Pinho-Ribeiro FA, Richardson Q, Jackson R, Chiu IM, Benoist C, Stevens B, Trejo JL, Mathis D. The meninges host a unique compartment of regulatory T cells that bulwarks adult hippocampal neurogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599387. [PMID: 38948783 PMCID: PMC11212874 DOI: 10.1101/2024.06.17.599387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Our knowledge about the meningeal immune system has recently burgeoned, particularly our understanding of how innate and adaptive effector cells are mobilized to meet brain challenges. However, information on how meningeal immunocytes guard brain homeostasis in healthy individuals remains sparse. This study highlights the heterogeneous and polyfunctional regulatory-T (Treg) cell compartment in the meninges. A Treg subtype specialized in controlling Th1-cell responses and another known to control responses in B-cell follicles were substantial components of this compartment, foretelling that punctual Treg-cell ablation rapidly unleashed interferon-gamma production by meningeal lymphocytes, unlocked their access to the brain parenchyma, and altered meningeal B-cell profiles. Distally, the hippocampus assumed a reactive state, with morphological and transcriptional changes in multiple glial-cell types; within the dentate gyrus, neural stem cells showed exacerbated death and desisted from further differentiation, associated with inhibition of spatial-reference memory. Thus, meningeal Treg cells are a multifaceted bulwark to brain homeostasis at steady-state. One sentence summary A distinct population of regulatory T cells in the murine meninges safeguards homeostasis by keeping local interferon-γ-producing lymphocytes in check, thereby preventing their invasion of the parenchyma, activation of hippocampal glial cells, death of neural stem cells, and memory decay.
Collapse
|
25
|
Deng T, Wang Z, Geng Q, Wang Z, Jiao Y, Diao W, Xu J, Deng T, Luo J, Tao Q, Xiao C. Methylation of T and B Lymphocytes in Autoimmune Rheumatic Diseases. Clin Rev Allergy Immunol 2024; 66:401-422. [PMID: 39207646 DOI: 10.1007/s12016-024-09003-4] [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] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
The role of abnormal epigenetic modifications, particularly DNA methylation, in the pathogenesis of autoimmune rheumatic diseases (ARDs) has garnered increasing attention. Lymphocyte dysfunction is a significant contributor to the pathogenesis of ARDs. Methylation is crucial for maintaining normal immune system function, and aberrant methylation can hinder lymphocyte differentiation, resulting in functional abnormalities that disrupt immune tolerance, leading to the excessive expression of inflammatory cytokines, thereby exacerbating the onset and progression of ARDs. Recent studies suggest that methylation-related factors have the potential to serve as biomarkers for monitoring the activity of ARDs. This review summarizes the current state of research on the impact of DNA and RNA methylation on the development, differentiation, and function of T and B cells and examines the progress of these epigenetic modifications in studies of six specific ARDs: systemic lupus erythematosus, rheumatoid arthritis, Sjögren's syndrome, systemic sclerosis, juvenile idiopathic arthritis, and ankylosing spondylitis. Additionally, we propose that exploring the interplay between RNA methylation and DNA methylation may represent a novel direction for understanding the pathogenesis of ARDs and developing novel treatment strategies.
Collapse
Affiliation(s)
- Tiantian Deng
- Beijing University of Chinese Medicine, School of Clinical Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zihan Wang
- Beijing University of Chinese Medicine, School of Clinical Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Qishun Geng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Zhaoran Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Yi Jiao
- Beijing University of Chinese Medicine, School of Clinical Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wenya Diao
- Beijing University of Chinese Medicine, School of Clinical Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jiahe Xu
- China-Japan Friendship Hospital, Peking University, Beijing, 100029, China
| | - Tingting Deng
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jing Luo
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Qingwen Tao
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
26
|
Watanabe M, Matsui A, Awata N, Nagafuchi A, Kawazoe M, Harada Y, Ito M. Differences in the characteristics and functions of brain and spinal cord regulatory T cells. J Neuroinflammation 2024; 21:146. [PMID: 38824594 PMCID: PMC11143704 DOI: 10.1186/s12974-024-03144-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024] Open
Abstract
T cells play an important role in the acquired immune response, with regulatory T cells (Tregs) serving as key players in immune tolerance. Tregs are found in nonlymphoid and damaged tissues and are referred to as "tissue Tregs". They have tissue-specific characteristics and contribute to immunomodulation, homeostasis, and tissue repair through interactions with tissue cells. However, important determinants of Treg tissue specificity, such as antigen specificity, tissue environment, and pathology, remain unclear. In this study, we analyzed Tregs in the central nervous system of mice with ischemic stroke and experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. The gene expression pattern of brain Tregs in the EAE model was more similar to that of ischemic stroke Tregs in the brain than to that of spinal cord Tregs. In addition, most T-cell receptors (TCRs) with high clonality were present in both the brain and spinal cord. Furthermore, Gata3+ and Rorc+ Tregs expressed TCRs recognizing MOG in the spinal cord, suggesting a tissue environment conducive to Rorc expression. Tissue-specific chemokine/chemokine receptor interactions in the spinal cord and brain influenced Treg localization. Finally, spinal cord- or brain-derived Tregs had greater anti-inflammatory capacities in EAE mice, respectively. Taken together, these findings suggest that the tissue environment, rather than pathogenesis or antigen specificity, is the primary determinant of the tissue-specific properties of Tregs. These findings may contribute to the development of novel therapies to suppress inflammation through tissue-specific Treg regulation.
Collapse
Affiliation(s)
- Mahiro Watanabe
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Ako Matsui
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Natsumi Awata
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Ayame Nagafuchi
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Mio Kawazoe
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yoshihiro Harada
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Minako Ito
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
27
|
Camerini E, Amsen D, Kater AP, Peters FS. The complexities of T-cell dysfunction in chronic lymphocytic leukemia. Semin Hematol 2024; 61:163-171. [PMID: 38782635 DOI: 10.1053/j.seminhematol.2024.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/13/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell malignancy characterized by profound alterations and defects in the T-cell compartment. This observation has gained renewed interest as T-cell treatment strategies, which are successfully applied in more aggressive B-cell malignancies, have yielded disappointing results in CLL. Despite ongoing efforts to understand and address the observed T-cell defects, the exact mechanisms and nature underlying this dysfunction remain largely unknown. In this review, we examine the supporting signals from T cells to CLL cells in the lymph node niche, summarize key findings on T-cell functional defects, delve into potential underlying causes, and explore novel strategies for reversing these deficiencies. Our goal is to identify strategies aimed at resolving CLL-induced T-cell dysfunction which, in the future, will enhance the efficacy of autologous T-cell-based therapies for CLL patients.
Collapse
Affiliation(s)
- Elena Camerini
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Derk Amsen
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Landsteiner Laboratory for Blood Cell Research at Sanquin, Amsterdam, The Netherlands
| | - Arnon P Kater
- Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Fleur S Peters
- Department of Experimental Immunology, Amsterdam UMC, Amsterdam, The Netherlands; Department of Hematology, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Abstract
The brain, long thought to be isolated from the peripheral immune system, is increasingly recognized to be integrated into a systemic immunological network. These conduits of immune-brain interaction and immunosurveillance processes necessitate the presence of complementary immunoregulatory mechanisms, of which brain regulatory T cells (Treg cells) are likely a key facet. Treg cells represent a dynamic population in the brain, with continual influx, specialization to a brain-residency phenotype and relatively rapid displacement by newly incoming cells. In addition to their functions in suppressing adaptive immunity, an emerging view is that Treg cells in the brain dampen down glial reactivity in response to a range of neurological insults, and directly assist in multiple regenerative and reparative processes during tissue pathology. The utility and malleability of the brain Treg cell population make it an attractive therapeutic target across the full spectrum of neurological conditions, ranging from neuroinflammatory to neurodegenerative and even psychiatric diseases. Therapeutic modalities currently under intense development include Treg cell therapy, IL-2 therapy to boost Treg cell numbers and multiple innovative approaches to couple these therapeutics to brain delivery mechanisms for enhanced potency. Here we review the state of the art of brain Treg cell knowledge together with the potential avenues for future integration into medical practice.
Collapse
Affiliation(s)
- Adrian Liston
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Emanuela Pasciuto
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- Center for Molecular Neurology, VIB, Antwerp, Belgium.
| | - Denise C Fitzgerald
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK.
| | - Lidia Yshii
- Department of Neurosciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Zhang YS, Chen YQ. Dysfunctional regulatory T cell: May be an obstacle to immunotherapy in cardiovascular diseases. Biomed Pharmacother 2024; 173:116359. [PMID: 38430633 DOI: 10.1016/j.biopha.2024.116359] [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: 12/06/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
Inflammatory responses are linked to cardiovascular diseases (CVDs) in various forms. Tregs, members of CD4+ T cells, play important roles in regulating immune system and suppressing inflammatory response, thus contributing to maintaining immune homeostasis. However, Tregs exert their powerful suppressive function relying on the stable phenotype and function. The stability of Tregs primarily depends on the FOXP3 (Forkhead box P3) expression and epigenetic regulation. Although Tregs are quite stable under physiological conditions, prolonged exposure to inflammatory cues, Tregs may lose suppressive function and require proinflammatory phenotype, namely plastic Tregs or ex-Tregs. There are extensive researches have established the beneficial role of Tregs in CVDs. Nevertheless, the potential risks of dysfunctional Tregs lack deep research. Anti-inflammatory and immunological modulation have been hotspots in the treatment of CVDs. Tregs are appealing because of their crucial role in resolving inflammation and promoting tissue repair. If alleviating inflammatory response through modulating Tregs could be a new therapeutic strategy for CVDs, the next step to consider is how to prevent the formation of dysfunctional Tregs or reverse detrimental Tregs to normal phenotype.
Collapse
Affiliation(s)
- Yu-Sha Zhang
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Hunan, China
| | - Ya-Qin Chen
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Hunan, China.
| |
Collapse
|
30
|
Alvarez F, Liu Z, Bay A, Piccirillo CA. Deciphering the developmental trajectory of tissue-resident Foxp3 + regulatory T cells. Front Immunol 2024; 15:1331846. [PMID: 38605970 PMCID: PMC11007185 DOI: 10.3389/fimmu.2024.1331846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/14/2024] [Indexed: 04/13/2024] Open
Abstract
Foxp3+ TREG cells have been at the focus of intense investigation for their recognized roles in preventing autoimmunity, facilitating tissue recuperation following injury, and orchestrating a tolerance to innocuous non-self-antigens. To perform these critical tasks, TREG cells undergo deep epigenetic, transcriptional, and post-transcriptional changes that allow them to adapt to conditions found in tissues both at steady-state and during inflammation. The path leading TREG cells to express these tissue-specialized phenotypes begins during thymic development, and is further driven by epigenetic and transcriptional modifications following TCR engagement and polarizing signals in the periphery. However, this process is highly regulated and requires TREG cells to adopt strategies to avoid losing their regulatory program altogether. Here, we review the origins of tissue-resident TREG cells, from their thymic and peripheral development to the transcriptional regulators involved in their tissue residency program. In addition, we discuss the distinct signalling pathways that engage the inflammatory adaptation of tissue-resident TREG cells, and how they relate to their ability to recognize tissue and pathogen-derived danger signals.
Collapse
Affiliation(s)
- Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
| | - Zhiyang Liu
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
| | - Alexandre Bay
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
| |
Collapse
|
31
|
Wachtendorf S, Jonin F, Ochel A, Heinrich F, Westendorf AM, Tiegs G, Neumann K. The ST2 + Treg/amphiregulin axis protects from immune-mediated hepatitis. Front Immunol 2024; 15:1351405. [PMID: 38571949 PMCID: PMC10987816 DOI: 10.3389/fimmu.2024.1351405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction The alarmin IL-33 has been implicated in the pathology of immune-mediated liver diseases. IL-33 activates regulatory T cells (Tregs) and type 2 innate lymphoid cells (ILC2s) expressing the IL-33 receptor ST2. We have previously shown that endogenous IL-33/ST2 signaling activates ILC2s that aggravate liver injury in murine immune-mediated hepatitis. However, treatment of mice with exogenous IL-33 before induction of hepatitis ameliorated disease severity. Since IL-33 induces expression of amphiregulin (AREG) crucial for Treg function, we investigated the immunoregulatory role of the ST2+ Treg/AREG axis in immune-mediated hepatitis. Methods C57BL/6, ST2-deficient (Il1rl1-/-) and Areg-/- mice received concanavalin A to induce immune-mediated hepatitis. Foxp3Cre+ x ST2fl/fl mice were pre-treated with IL-33 before induction of immune-mediated hepatitis. Treg function was assessed by adoptive transfer experiments and suppression assays. The effects of AREG and IL-33 on ST2+ Tregs and ILC2s were investigated in vitro. Immune cell phenotype was analyzed by flow cytometry. Results and discussion We identified IL-33-responsive ST2+ Tregs as an effector Treg subset in the murine liver, which was highly activated in immune-mediated hepatitis. Lack of endogenous IL-33 signaling in Il1rl1-/- mice aggravated disease pathology. This was associated with reduced Treg activation. Adoptive transfer of exogenous IL-33-activated ST2+ Tregs before induction of hepatitis suppressed inflammatory T-cell responses and ameliorated disease pathology. We further showed increased expression of AREG by hepatic ST2+ Tregs and ILC2s in immune-mediated hepatitis. Areg-/- mice developed more severe liver injury, which was associated with enhanced ILC2 activation and less ST2+ Tregs in the inflamed liver. Exogenous AREG suppressed ILC2 cytokine expression and enhanced ST2+ Treg activation in vitro. In addition, Tregs from Areg-/- mice were impaired in their capacity to suppress CD4+ T-cell activation in vitro. Moreover, application of exogenous IL-33 before disease induction did not protect Foxp3Cre+ x ST2fl/fl mice lacking ST2+ Tregs from immune-mediated hepatitis. In summary, we describe an immunoregulatory role of the ST2+ Treg/AREG axis in immune-mediated hepatitis, in which AREG suppresses the activation of hepatic ILC2s while maintaining ST2+ Tregs and reinforcing their immunosuppressive capacity in liver inflammation.
Collapse
Affiliation(s)
- Selina Wachtendorf
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fitriasari Jonin
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Aaron Ochel
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Heinrich
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Astrid M. Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Neumann
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
32
|
Rath S, Hawsawi YM, Alzahrani F, Khan MI. Epigenetic regulation of inflammation: The metabolomics connection. Semin Cell Dev Biol 2024; 154:355-363. [PMID: 36127262 DOI: 10.1016/j.semcdb.2022.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/12/2022] [Accepted: 09/12/2022] [Indexed: 10/14/2022]
Abstract
Epigenetic factors are considered the regulator of complex machinery behind inflammatory disorders and significantly contributed to the expression of inflammation-associated genes. Epigenetic modifications modulate variation in the expression pattern of target genes without affecting the DNA sequence. The current knowledge of epigenetic research focused on their role in the pathogenesis of various inflammatory diseases that causes morbidity and mortality worldwide. Inflammatory diseases are categorized as acute and chronic based on the disease severity and are regulated by the expression pattern of various genes. Hence, understanding the role of epigenetic modifications during inflammation progression will contribute to the disease outcomes and therapeutic approaches. This review also focuses on the metabolomics approach associated with the study of inflammatory disorders. Inflammatory responses and metabolic regulation are highly integrated and various advanced techniques are adopted to study the metabolic signature molecules. Here we discuss several metabolomics approaches used to link inflammatory disorders and epigenetic changes. We proposed that deciphering the mechanism behind the inflammation-metabolism loop may have immense importance in biomarkers research and may act as a principal component in drug discovery as well as therapeutic applications.
Collapse
Affiliation(s)
- Suvasmita Rath
- Center of Environment, Climate Change and Public Health, Utkal University, Vani Vihar, Bhubaneswar 751004, Odisha, India
| | - Yousef M Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 40047, Jeddah 21499, Saudi Arabia; College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia.
| | - Faisal Alzahrani
- Department of Biochemistry, King Abdulaziz University (KAU), Jeddah 21577, Saudi Arabia; Embryonic Stem Cells Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Imran Khan
- Department of Biochemistry, King Abdulaziz University (KAU), Jeddah 21577, Saudi Arabia; Centre of Artificial Intelligence for Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
33
|
Zhao Y, Nicholson L, Wang H, Qian YW, Hawthorne WJ, Jimenez-Vera E, Gloss BS, Lai J, Thomas A, Chew YV, Burns H, Zhang GY, Wang YM, Rogers NM, Zheng G, Yi S, Alexander SI, O’Connell PJ, Hu M. Intragraft memory-like CD127hiCD4+Foxp3+ Tregs maintain transplant tolerance. JCI Insight 2024; 9:e169119. [PMID: 38516885 PMCID: PMC11063946 DOI: 10.1172/jci.insight.169119] [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: 02/14/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) play an essential role in suppressing transplant rejection, but their role within the graft and heterogeneity in tolerance are poorly understood. Here, we compared phenotypic and transcriptomic characteristics of Treg populations within lymphoid organs and grafts in an islet xenotransplant model of tolerance. We showed Tregs were essential for tolerance induction and maintenance. Tregs demonstrated heterogeneity within the graft and lymphoid organs of tolerant mice. A subpopulation of CD127hi Tregs with memory features were found in lymphoid organs, presented in high proportions within long-surviving islet grafts, and had a transcriptomic and phenotypic profile similar to tissue Tregs. Importantly, these memory-like CD127hi Tregs were better able to prevent rejection by effector T cells, after adoptive transfer into secondary Rag-/- hosts, than naive Tregs or unselected Tregs from tolerant mice. Administration of IL-7 to the CD127hi Treg subset was associated with a strong activation of phosphorylation of STAT5. We proposed that memory-like CD127hi Tregs developed within the draining lymph node and underwent further genetic reprogramming within the graft toward a phenotype that had shared characteristics with other tissue or tumor Tregs. These findings suggested that engineering Tregs with these characteristics either in vivo or for adoptive transfer could enhance transplant tolerance.
Collapse
Affiliation(s)
| | | | - Hannah Wang
- Centre for Transplant and Renal Research and
| | - Yi Wen Qian
- Centre for Transplant and Renal Research and
| | | | | | - Brian S. Gloss
- Scientific Platforms, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Joey Lai
- Scientific Platforms, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | | | - Yi Vee Chew
- Centre for Transplant and Renal Research and
| | | | - Geoff Y. Zhang
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | - Natasha M. Rogers
- Centre for Transplant and Renal Research and
- Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | | | - Shounan Yi
- Centre for Transplant and Renal Research and
| | - Stephen I. Alexander
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | | | - Min Hu
- Centre for Transplant and Renal Research and
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
34
|
Becker M, Dirschl SM, Scherm MG, Serr I, Daniel C. Niche-specific control of tissue function by regulatory T cells-Current challenges and perspectives for targeting metabolic disease. Cell Metab 2024; 36:229-239. [PMID: 38218187 DOI: 10.1016/j.cmet.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/15/2023] [Indexed: 01/15/2024]
Abstract
Tissue regulatory T cells (Tregs) exert pivotal functions in both immune and metabolic regulation, maintaining local tissue homeostasis, integrity, and function. Accordingly, Tregs play a crucial role in controlling obesity-induced inflammation and supporting efficient muscle function and repair. Depending on the tissue context, Tregs are characterized by unique transcriptomes, growth, and survival factors and T cell receptor (TCR) repertoires. This functional specialization offers the potential to selectively target context-specific Treg populations, tailoring therapeutic strategies to specific niches, thereby minimizing potential side effects. Here, we discuss challenges and perspectives for niche-specific Treg targeting, which holds promise for highly efficient and precise medical interventions to combat metabolic disease.
Collapse
Affiliation(s)
- Maike Becker
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany
| | - Sandra M Dirschl
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany
| | - Martin G Scherm
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany
| | - Isabelle Serr
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany
| | - Carolin Daniel
- Research Division Type 1 Diabetes Immunology, Helmholtz Diabetes Center at Helmholtz Zentrum München, 80939 Munich, Germany; Deutsches Zentrum für Diabetesforschung (DZD), 85764 Munich, Germany; Division of Clinical Pharmacology, Department of Medicine IV, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
35
|
Delacher M, Schmidleithner L, Simon M, Stüve P, Sanderink L, Hotz-Wagenblatt A, Wuttke M, Schambeck K, Ruhland B, Hofmann V, Bittner S, Ritter U, Pant A, Helbich SS, Voss M, Lemmermann NA, Bessiri-Schake L, Bohn T, Eigenberger A, Menevse AN, Gebhard C, Strieder N, Abken H, Rehli M, Huehn J, Beckhove P, Hehlgans T, Junger H, Geissler EK, Prantl L, Werner JM, Schmidl C, Brors B, Imbusch CD, Feuerer M. The effector program of human CD8 T cells supports tissue remodeling. J Exp Med 2024; 221:e20230488. [PMID: 38226976 PMCID: PMC10791561 DOI: 10.1084/jem.20230488] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/19/2023] [Accepted: 12/06/2023] [Indexed: 01/17/2024] Open
Abstract
CD8 T lymphocytes are classically viewed as cytotoxic T cells. Whether human CD8 T cells can, in parallel, induce a tissue regeneration program is poorly understood. Here, antigen-specific assay systems revealed that human CD8 T cells not only mediated cytotoxicity but also promoted tissue remodeling. Activated CD8 T cells could produce the epidermal growth factor receptor (EGFR)-ligand amphiregulin (AREG) and sensitize epithelial cells for enhanced regeneration potential. Blocking the EGFR or the effector cytokines IFN-γ and TNF could inhibit tissue remodeling. This regenerative program enhanced tumor spheroid and stem cell-mediated organoid growth. Using single-cell gene expression analysis, we identified an AREG+, tissue-resident CD8 T cell population in skin and adipose tissue from patients undergoing abdominal wall or abdominoplasty surgery. These tissue-resident CD8 T cells showed a strong TCR clonal relation to blood PD1+TIGIT+ CD8 T cells with tissue remodeling abilities. These findings may help to understand the complex CD8 biology in tumors and could become relevant for the design of therapeutic T cell products.
Collapse
Affiliation(s)
- Michael Delacher
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Lisa Schmidleithner
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Malte Simon
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Philipp Stüve
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Lieke Sanderink
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Agnes Hotz-Wagenblatt
- Core Facility Omics IT and Data Management, German Cancer Research Center, Heidelberg, Germany
| | - Marina Wuttke
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Kathrin Schambeck
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Brigitte Ruhland
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Veronika Hofmann
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Sebastian Bittner
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Uwe Ritter
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Asmita Pant
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Sara Salome Helbich
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Morten Voss
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Niels A. Lemmermann
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
- Institute of Virology, University Medical Center Mainz, Mainz, Germany
- Institute of Virology, University of Bonn, Bonn, Germany
| | - Lisa Bessiri-Schake
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Toszka Bohn
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Andreas Eigenberger
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Ayse Nur Menevse
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Interventional Immunology, University Regensburg, Regensburg, Germany
| | | | | | - Hinrich Abken
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Genetic Immunotherapy, University Regensburg, Regensburg, Germany
| | - Michael Rehli
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Hannover Medical School, Hannover, Germany
- RESIST, Cluster of Excellence 2155, Hannover Medical School, Hannover, Germany
| | - Philipp Beckhove
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Interventional Immunology, University Regensburg, Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Hehlgans
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Henrik Junger
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Edward K. Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand- and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jens M. Werner
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | | | - Benedikt Brors
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Faculty of Medicine Heidelberg, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center, Heidelberg, Germany
- National Center for Tumor Diseases, Heidelberg, Germany
- German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Charles D. Imbusch
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
- Division of Applied Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Markus Feuerer
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| |
Collapse
|
36
|
Muñoz-Rojas AR, Wang G, Benoist C, Mathis D. Adipose-tissue regulatory T cells are a consortium of subtypes that evolves with age and diet. Proc Natl Acad Sci U S A 2024; 121:e2320602121. [PMID: 38227656 PMCID: PMC10823167 DOI: 10.1073/pnas.2320602121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024] Open
Abstract
Foxp3+CD4+ regulatory T (Treg) cells found within tissues regulate local immunity, inflammation, and homeostasis. Tregs in epididymal visceral adipose tissue (eVAT) are critical regulators of local and systemic inflammation and metabolism. During aging and under obesogenic conditions, eVAT Tregs undergo transcriptional and phenotypic changes and are important for containing inflammation and normalizing metabolic indices. We have employed single-cell RNA sequencing, single-cell Tra and Trb sequencing, adoptive transfers, photoconvertible mice, cellular interaction analyses, and in vitro cultures to dissect the evolving heterogeneity of eVAT Tregs with aging and obesity. Distinct Treg subtypes with distinguishable gene expression profiles and functional roles were enriched at differing ages and with differing diets. Like those in lean mice, eVAT Tregs in obese mice were not primarily recruited from the circulation but instead underwent local expansion and had a distinct and diversified T cell receptor repertoire. The different eVAT-Treg subtypes were specialized in different functions; for example, the subtypes enriched in lean, but not obese, mice suppressed adipogenesis. The existence of functionally divergent eVAT-Treg subtypes in response to obesogenic conditions presents possibilities for precision therapeutics in the context of obesity.
Collapse
Affiliation(s)
| | - Gang Wang
- Department of Immunology, Harvard Medical School, Boston, MA02115
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
37
|
Georgiev P, Benamar M, Han S, Haigis MC, Sharpe AH, Chatila TA. Regulatory T cells in dominant immunologic tolerance. J Allergy Clin Immunol 2024; 153:28-41. [PMID: 37778472 PMCID: PMC10842646 DOI: 10.1016/j.jaci.2023.09.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023]
Abstract
Regulatory T cells expressing the transcription factor forkhead box protein 3 mediate peripheral immune tolerance both to self-antigens and to the commensal flora. Their defective function due to inborn errors of immunity or acquired insults is associated with a broad range of autoimmune and immune dysregulatory diseases. Although their function in suppressing autoimmunity and enforcing commensalism is established, a broader role for regulatory T cells in tissue repair and metabolic regulation has emerged, enabled by unique programs of tissue adaptability and specialization. In this review, we focus on the myriad roles played by regulatory T cells in immunologic tolerance and host homeostasis and the potential to harness these cells in novel therapeutic approaches to human diseases.
Collapse
Affiliation(s)
- Peter Georgiev
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Mass; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Mass
| | - Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - SeongJun Han
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Mass; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Mass
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Mass
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Mass
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
38
|
Yilmazer A, Zevla DM, Malmkvist R, Rodríguez CAB, Undurraga P, Kirgin E, Boernert M, Voehringer D, Kershaw O, Schlenner S, Kretschmer K. Selective ablation of thymic and peripheral Foxp3 + regulatory T cell development. Front Immunol 2023; 14:1298938. [PMID: 38164128 PMCID: PMC10757929 DOI: 10.3389/fimmu.2023.1298938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Foxp3+ regulatory T (Treg) cells of thymic (tTreg) and peripheral (pTreg) developmental origin are thought to synergistically act to ensure immune homeostasis, with self-reactive tTreg cells primarily constraining autoimmune responses. Here we exploited a Foxp3-dependent reporter with thymus-specific GFP/Cre activity to selectively ablate either tTreg (ΔtTreg) or pTreg (ΔpTreg) cell development, while sparing the respective sister populations. We found that, in contrast to the tTreg cell behavior in ΔpTreg mice, pTreg cells acquired a highly activated suppressor phenotype and replenished the Treg cell pool of ΔtTreg mice on a non-autoimmune C57BL/6 background. Despite the absence of tTreg cells, pTreg cells prevented early mortality and fatal autoimmunity commonly observed in Foxp3-deficient models of complete Treg cell deficiency, and largely maintained immune tolerance even as the ΔtTreg mice aged. However, only two generations of backcrossing to the autoimmune-prone non-obese diabetic (NOD) background were sufficient to cause severe disease lethality associated with different, partially overlapping patterns of organ-specific autoimmunity. This included a particularly severe form of autoimmune diabetes characterized by an early onset and abrogation of the sex bias usually observed in the NOD mouse model of human type 1 diabetes. Genetic association studies further allowed us to define a small set of autoimmune risk loci sufficient to promote β cell autoimmunity, including genes known to impinge on Treg cell biology. Overall, these studies show an unexpectedly high functional adaptability of pTreg cells, emphasizing their important role as mediators of bystander effects to ensure self-tolerance.
Collapse
Affiliation(s)
- Acelya Yilmazer
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Dimitra Maria Zevla
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Rikke Malmkvist
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Carlos Alejandro Bello Rodríguez
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Pablo Undurraga
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Emre Kirgin
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Marie Boernert
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - David Voehringer
- Department of Infection Biology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Olivia Kershaw
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Susan Schlenner
- KU Leuven-University of Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Karsten Kretschmer
- Molecular and Cellular Immunology/Immune Regulation, Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
39
|
Griffith JW, Faustino LD, Cottrell VI, Nepal K, Hariri LP, Chiu RSY, Jones MC, Julé A, Gabay C, Luster AD. Regulatory T cell-derived IL-1Ra suppresses the innate response to respiratory viral infection. Nat Immunol 2023; 24:2091-2107. [PMID: 37945820 PMCID: PMC11887468 DOI: 10.1038/s41590-023-01655-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 09/15/2023] [Indexed: 11/12/2023]
Abstract
Regulatory T (Treg) cell modulation of adaptive immunity and tissue homeostasis is well described; however, less is known about Treg cell-mediated regulation of the innate immune response. Here we show that deletion of ST2, the receptor for interleukin (IL)-33, on Treg cells increased granulocyte influx into the lung and increased cytokine production by innate lymphoid and γδ T cells without alteration of adaptive immunity to influenza. IL-33 induced high levels of the interleukin-1 receptor antagonist (IL-1Ra) in ST2+ Treg cells and deletion of IL-1Ra in Treg cells increased granulocyte influx into the lung. Treg cell-specific deletion of ST2 or IL-1Ra improved survival to influenza, which was dependent on IL-1. Adventitial fibroblasts in the lung expressed high levels of the IL-1 receptor and their chemokine production was suppressed by Treg cell-produced IL-1Ra. Thus, we define a new pathway where IL-33-induced IL-1Ra production by tissue Treg cells suppresses IL-1-mediated innate immune responses to respiratory viral infection.
Collapse
Affiliation(s)
- Jason W Griffith
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lucas D Faustino
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Victoria I Cottrell
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keshav Nepal
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lida P Hariri
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca Suet-Yan Chiu
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael C Jones
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amélie Julé
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Cem Gabay
- Division of Rheumatology, University Hospitals of Geneva and University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Zhong G, Wu D, Chen H, Yan L, Xiang Q, Liu Y, Wang T. Multi-omics Analysis of Prognostic Significance and Immune Infiltration of FASTK Family Members in Kidney Renal Clear Cell Carcinoma. Evol Bioinform Online 2023; 19:11769343231212078. [PMID: 38033663 PMCID: PMC10683404 DOI: 10.1177/11769343231212078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/18/2023] [Indexed: 12/02/2023] Open
Abstract
Objective The Fas-activated serine/threonine kinase (FASTK) family of proteins has been recently found to be able to regulate mitochondrial gene expression post-transcriptionally. Nonetheless, there is a paucity of study about the role of the FASTK family in kidney renal clear cell carcinoma (KIRC). This study was conducted to explore the correlation of FASTK family genes with expression, prognosis, and immune infiltration in KIRC. Methods We collected the data from the UALCAN, GeneMANIA, STRING, CancerSEA, cBioPortal, Kaplan-Meier plotter, GEPIA, TISIDB and TIMER databases to evaluate the genetic alterations, differential expression, prognostic significance, and immune cell infiltration of FASTKs in patients with KIRC. Results In tumor tissues of KIRC, the mRNA expression level of FASTK and TBRG4 was elevated, whereas that of FASTKD1, FASTKD2, and FASTKD5 was lowered compared with normal tissues (P < .05). Patients with KIRC and high FASTK and Transforming growth factor β regulator 4 (TBRG4) expression had worse overall survival (OS) and disease specific survival (DFS), while those with lower expression of FASTKD2/3/5 had worse outcomes. FASTK was positively correlated with DNA damage. FASTKD1 was positively related to differentiation. FASTKD2 was inversely related to proliferation and FASTKD5 was inversely related to invasion and EMT in KIRC cells. FASTK expression in KIRC was inversely linked to the presence of several immune cells including Tgd, macrophages, Tcm, and Mast cells (P < .05). Conclusions Our research provided fresh insight and in-depth analysis to the selection of prognostic biological markers of FASTK family members in KIRC.
Collapse
Affiliation(s)
- Guanghui Zhong
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Dali Wu
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Haiping Chen
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Lingfei Yan
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Qi Xiang
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yufeng Liu
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Tao Wang
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Liang Y, Wang L, Ma P, Ju D, Zhao M, Shi Y. Enhancing anti-tumor immune responses through combination therapies: epigenetic drugs and immune checkpoint inhibitors. Front Immunol 2023; 14:1308264. [PMID: 38077327 PMCID: PMC10704038 DOI: 10.3389/fimmu.2023.1308264] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Epigenetic mechanisms are processes that affect gene expression and cellular functions without involving changes in the DNA sequence. This abnormal or unstable expression of genes regulated by epigenetics can trigger cancer and other various diseases. The immune cells involved in anti-tumor responses and the immunogenicity of tumors may also be affected by epigenomic changes. This holds significant implications for the development and application of cancer immunotherapy, epigenetic therapy, and their combined treatments in the fight against cancer. We provide an overview of recent research literature focusing on how epigenomic changes in immune cells influence immune cell behavior and function, as well as the immunogenicity of cancer cells. And the combined utilization of epigenetic medications with immune checkpoint inhibitors that focus on immune checkpoint molecules [e.g., Programmed Death 1 (PD-1), Cytotoxic T-Lymphocyte-Associated Protein 4 (CTLA-4), T cell Immunoglobulin and Mucin Domain (TIM-3), Lymphocyte Activation Gene-3 (LAG-3)] present in immune cells and stromal cells associated with tumors. We highlight the potential of small-molecule inhibitors targeting epigenetic regulators to amplify anti-tumor immune responses. Moreover, we discuss how to leverage the intricate relationship between cancer epigenetics and cancer immunology to create treatment regimens that integrate epigenetic therapies with immunotherapies.
Collapse
Affiliation(s)
- Ying Liang
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Lingling Wang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan Wuchang Hospital, Wuhan, China
| | - Peijun Ma
- Clinical Laboratory, Shanghai Mental Health Center, Shanghai, China
| | - Dongen Ju
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Minggao Zhao
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yun Shi
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| |
Collapse
|
42
|
Abstract
Regulatory T (Treg) cells maintain immune tolerance to allergens at the environmental interfaces in the airways, skin and gut, marshalling in the process distinct immune regulatory circuits operative in the respective tissues. Treg cells are coordinately mobilized with allergic effector mechanisms in the context of a tissue-protective allergic inflammatory response against parasites, toxins and potentially harmful allergens, serving to both limit the inflammation and promote local tissue repair. Allergic diseases are associated with subverted Treg cell responses whereby a chronic allergic inflammatory environment can skew Treg cells toward pathogenic phenotypes that both perpetuate and aggravate disease. Interruption of Treg cell subversion in chronic allergic inflammatory conditions may thus provide novel therapeutic strategies by re-establishing effective immune regulation.
Collapse
Affiliation(s)
- Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Qian Chen
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Monica Martinez-Blanco
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Lead Contact, USA.
| |
Collapse
|
43
|
Knoedler S, Knoedler L, Kauke-Navarro M, Rinkevich Y, Hundeshagen G, Harhaus L, Kneser U, Pomahac B, Orgill DP, Panayi AC. Regulatory T cells in skin regeneration and wound healing. Mil Med Res 2023; 10:49. [PMID: 37867188 PMCID: PMC10591349 DOI: 10.1186/s40779-023-00484-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 10/04/2023] [Indexed: 10/24/2023] Open
Abstract
As the body's integumentary system, the skin is vulnerable to injuries. The subsequent wound healing processes aim to restore dermal and epidermal integrity and functionality. To this end, multiple tissue-resident cells and recruited immune cells cooperate to efficiently repair the injured tissue. Such temporally- and spatially-coordinated interplay necessitates tight regulation to prevent collateral damage such as overshooting immune responses and excessive inflammation. In this context, regulatory T cells (Tregs) hold a key role in balancing immune homeostasis and mediating cutaneous wound healing. A comprehensive understanding of Tregs' multifaceted field of activity may help decipher wound pathologies and, ultimately, establish new treatment modalities. Herein, we review the role of Tregs in orchestrating the regeneration of skin adnexa and catalyzing healthy wound repair. Further, we discuss how Tregs operate during fibrosis, keloidosis, and scarring.
Collapse
Affiliation(s)
- Samuel Knoedler
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, 06510, USA
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, 85764, Germany
| | - Leonard Knoedler
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Martin Kauke-Navarro
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, 85764, Germany
| | - Gabriel Hundeshagen
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, 67071, Germany
| | - Leila Harhaus
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, 67071, Germany
| | - Ulrich Kneser
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, 67071, Germany
| | - Bohdan Pomahac
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Dennis P Orgill
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Adriana C Panayi
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, 67071, Germany.
| |
Collapse
|
44
|
Braband KL, Nedwed AS, Helbich SS, Simon M, Beumer N, Brors B, Marini F, Delacher M. Using single-cell chromatin accessibility sequencing to characterize CD4+ T cells from murine tissues. Front Immunol 2023; 14:1232511. [PMID: 37908367 PMCID: PMC10613658 DOI: 10.3389/fimmu.2023.1232511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/29/2023] [Indexed: 11/02/2023] Open
Abstract
The Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) is a cutting-edge technology that enables researchers to assess genome-wide chromatin accessibility and to characterize cell type specific gene-regulatory programs. Recent technological progress allows for using this technology also on the single-cell level. In this article, we describe the whole value chain from the isolation of T cells from murine tissues to a complete bioinformatic analysis workflow. We start with methods for isolating scATAC-seq-ready CD4+ T cells from murine tissues such as visceral adipose tissue, skin, colon, and secondary lymphoid tissues such as the spleen. We describe the preparation of nuclei and quality control parameters during library preparation. Based on publicly available sequencing data that was generated using these protocols, we describe a step-by-step bioinformatic analysis pipeline for data pre-processing and downstream analysis. Our analysis workflow will follow the R-based bioinformatics framework ArchR, which is currently well established for scATAC-seq datasets. All in all, this work serves as a one-stop shop for generating and analyzing chromatin accessibility landscapes in T cells.
Collapse
Affiliation(s)
- Kathrin Luise Braband
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center Mainz, Mainz, Germany
| | - Annekathrin Silvia Nedwed
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Sara Salome Helbich
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center Mainz, Mainz, Germany
| | - Malte Simon
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Niklas Beumer
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Federico Marini
- Research Center for Immunotherapy (FZI), University Medical Center Mainz, Mainz, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Michael Delacher
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
45
|
Park SY, Yang H, Kim S, Yang J, Go H, Bae H. Alpha-Synuclein-Specific Regulatory T Cells Ameliorate Parkinson's Disease Progression in Mice. Int J Mol Sci 2023; 24:15237. [PMID: 37894917 PMCID: PMC10607030 DOI: 10.3390/ijms242015237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Parkinson's disease (PD) is a long-term neurodegenerative disease characterized by dopaminergic neuronal loss and the aggregation of alpha-synuclein (α-syn) in the brain. Cell therapy using regulatory T cells (Tregs) has therapeutic potential on PD progression in a mouse model; however, several challenges were associated with its applications. Here, we propose a strategy for α-syn specific Treg expansion (α-syn Treg). We presented α-syn to T cells via dendritic cells. This method increased the mobility of Tregs towards the site of abundant α-syn in vitro (p < 0.01; α-syn Tregs versus polyclonal Tregs (poly Tregs)) and in vivo. Consequently, α-syn Tregs showed noteworthy neuroprotective effects against motor function deficits (p < 0.05, p < 0.01; α-syn Tregs versus poly Tregs), dopaminergic neuronal loss (p < 0.001; α-syn Tregs versus poly Tregs), and α-syn accumulation (p < 0.05; α-syn Tregs versus poly Tregs) in MPTP-induced PD mice. Furthermore, the adoptive transfer of α-syn Tregs exerted immunosuppressive effects on activated microglia, especially pro-inflammatory microglia, in PD mice. Our findings suggest that α-syn presentation may provide a significant improvement in neuroprotective activities of Tregs and suggest the effective clinical application of Treg therapy in PD.
Collapse
Affiliation(s)
- Seon-Young Park
- Department of Science in Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (S.-Y.P.); (H.Y.); (S.K.)
| | - HyeJin Yang
- Department of Science in Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (S.-Y.P.); (H.Y.); (S.K.)
| | - Soyoung Kim
- Department of Science in Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (S.-Y.P.); (H.Y.); (S.K.)
| | - Juwon Yang
- Department of Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (J.Y.); (H.G.)
| | - Hyemin Go
- Department of Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (J.Y.); (H.G.)
| | - Hyunsu Bae
- Department of Science in Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (S.-Y.P.); (H.Y.); (S.K.)
- Department of Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (J.Y.); (H.G.)
| |
Collapse
|
46
|
Nedwed AS, Helbich SS, Braband KL, Volkmar M, Delacher M, Marini F. Using combined single-cell gene expression, TCR sequencing and cell surface protein barcoding to characterize and track CD4+ T cell clones from murine tissues. Front Immunol 2023; 14:1241283. [PMID: 37901204 PMCID: PMC10602882 DOI: 10.3389/fimmu.2023.1241283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/31/2023] [Indexed: 10/31/2023] Open
Abstract
Single-cell gene expression analysis using sequencing (scRNA-seq) has gained increased attention in the past decades for studying cellular transcriptional programs and their heterogeneity in an unbiased manner, and novel protocols allow the simultaneous measurement of gene expression, T-cell receptor clonality and cell surface protein expression. In this article, we describe the methods to isolate scRNA/TCR-seq-compatible CD4+ T cells from murine tissues, such as skin, spleen, and lymph nodes. We describe the processing of cells and quality control parameters during library preparation, protocols for multiplexing of samples, and strategies for sequencing. Moreover, we describe a step-by-step bioinformatic analysis pipeline from sequencing data generated using these protocols. This includes quality control, preprocessing of sequencing data and demultiplexing of individual samples. We perform quantification of gene expression and extraction of T-cell receptor alpha and beta chain sequences, followed by quality control and doublet detection, and methods for harmonization and integration of datasets. Next, we describe the identification of highly variable genes and dimensionality reduction, clustering and pseudotemporal ordering of data, and we demonstrate how to visualize the results with interactive and reproducible dashboards. We will combine different analytic R-based frameworks such as Bioconductor and Seurat, illustrating how these can be interoperable to optimally analyze scRNA/TCR-seq data of CD4+ T cells from murine tissues.
Collapse
Affiliation(s)
- Annekathrin Silvia Nedwed
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Sara Salome Helbich
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Kathrin Luise Braband
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Michael Volkmar
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), Mainz, Germany
| | - Michael Delacher
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Federico Marini
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
47
|
Guo S, Qian C, Li W, Zeng Z, Cai J, Luo Y. Modulation of Neuroinflammation: Advances in Roles and Mechanisms of the IL-33/ST2 Axis Involved in Ischemic Stroke. Neuroimmunomodulation 2023; 30:226-236. [PMID: 37729881 PMCID: PMC10614518 DOI: 10.1159/000533984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/02/2023] [Indexed: 09/22/2023] Open
Abstract
Interleukin (IL)-33 was initially recognized as a constituent of the IL-1 cytokine family in 2005. It exerts pleiotropic effects by regulating immune responses via its binding to the receptor ST2 (IL-33R). The IL-33/ST2 pathway has been linked to several inflammatory disorders. In human and rodents, the broad expression of IL-33 in spinal cord tissues and brain indicates its central nervous system-specific functions. Growing evidence supports the protective effects of the IL-33/ST2 pathway in ischemic stroke, along with a better understanding of the underlying mechanisms. IL-33 plays a crucial role in the regulation of the release of inflammatory molecules from glial cells in response to neuropathological lesions. Moreover, IL-33/ST2-mediated neuroprotection following cerebral ischemia may be linked to T-cell function, specifically regulatory T cells. Soluble ST2 (sST2) acts as a decoy receptor in the IL-33/ST2 axis, blocking IL-33 signaling through the membrane ST2 receptor. sST2 has also been identified as a potential inflammatory biomarker of ischemic stroke. Targeting sST2 specifically to eliminate its inhibition of the protective IL-33/ST2 pathway in ischemic brain tissues is a promising approach for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Shuang Guo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chengli Qian
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenfeng Li
- Department of Clinical Medicine, The Second Clinical College, Wuhan University, Wuhan, China
| | - Zhikun Zeng
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junlong Cai
- Clinical Trial Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Luo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
48
|
Yuan X, Jiang H, Fu D, Rech JC, Robida A, Rajanayake K, Yuan H, He M, Wen B, Sun D, Liu C, Chinnaswamy K, Stuckey JA, Paczesny S, Yang CY. Prophylactic Mitigation of Acute Graft versus Host Disease by Novel 2-(Pyrrolidin-1-ylmethyl)pyrrole-Based Stimulation-2 (ST2) Inhibitors. ACS Pharmacol Transl Sci 2023; 6:1275-1287. [PMID: 37705593 PMCID: PMC10496145 DOI: 10.1021/acsptsci.3c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Indexed: 09/15/2023]
Abstract
Hematopoietic cell transplantation (HCT) is a proven and potentially curable therapy for hematological malignancies and inherited hematological disease. The main risk of HCT is the development of graft versus host disease (GVHD) acquired in up to 50% of patients. Upregulation of soluble ST2 (sST2) is a key clinical biomarker for GVHD prognosis and was shown to be a potential therapeutic target for GVHD. Agents targeting sST2 to reduce the sST2 level after HCT have the potential to mitigate GVHD progression. Here, we report 32 (or XY52) as the lead ST2 inhibitor from our optimization campaign. XY52 had improved inhibitory activity and metabolic stability in vitro and in vivo. XY52 suppressed proinflammatory T-cell proliferation while increasing regulatory T cells in vitro. In a clinically relevant GVHD model, a 21-day prophylactic regimen of XY52 reduced plasma sST2 and IFN-γ levels and GVHD score and extended survival in mice. XY52 represented a significant improvement over our previous compound, iST2-1, and further optimization of XY52 is warranted. The small-molecule ST2 inhibitors can potentially be used as a biomarker-guided therapy for mitigating GVHD in future clinical applications.
Collapse
Affiliation(s)
- Xinrui Yuan
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Hua Jiang
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Denggang Fu
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Jason C. Rech
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Aaron Robida
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Krishani Rajanayake
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hebao Yuan
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Miao He
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chen Liu
- Department
of Pathology, Yale University, New Haven, Connecticut 06520, United States
| | - Krishnapriya Chinnaswamy
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jeanne A. Stuckey
- Michigan Center for Therapeutic Innovation, Department
of Internal
Medicine, Life Sciences Institute, Department of Pharmaceutical Sciences, College of
Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Sophie Paczesny
- Department
of Microbiology & Immunology, Medical
University of South Carolina, Charleston, South Carolina 29425-2503, United States
| | - Chao-Yie Yang
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
49
|
Sivasami P, Elkins C, Diaz-Saldana PP, Goss K, Peng A, Hamersky M, Bae J, Xu M, Pollack BP, Horwitz EM, Scharer CD, Seldin L, Li C. Obesity-induced dysregulation of skin-resident PPARγ + Treg cells promotes IL-17A-mediated psoriatic inflammation. Immunity 2023; 56:1844-1861.e6. [PMID: 37478855 PMCID: PMC10527179 DOI: 10.1016/j.immuni.2023.06.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/16/2023] [Accepted: 06/22/2023] [Indexed: 07/23/2023]
Abstract
Obesity is a major risk factor for psoriasis, but how obesity disrupts the regulatory mechanisms that keep skin inflammation in check is unclear. Here, we found that skin was enriched with a unique population of CD4+Foxp3+ regulatory T (Treg) cells expressing the nuclear receptor peroxisome proliferation-activated receptor gamma (PPARγ). PPARγ drove a distinctive transcriptional program and functional suppression of IL-17A+ γδ T cell-mediated psoriatic inflammation. Diet-induced obesity, however, resulted in a reduction of PPARγ+ skin Treg cells and a corresponding loss of control over IL-17A+ γδ T cell-mediated inflammation. Mechanistically, PPARγ+ skin Treg cells preferentially took up elevated levels of long-chain free fatty acids in obese mice, which led to cellular lipotoxicity, oxidative stress, and mitochondrial dysfunction. Harnessing the anti-inflammatory properties of these PPARγ+ skin Treg cells could have therapeutic potential for obesity-associated inflammatory skin diseases.
Collapse
Affiliation(s)
- Pulavendran Sivasami
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cody Elkins
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Pamela P Diaz-Saldana
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kyndal Goss
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Amy Peng
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael Hamersky
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jennifer Bae
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Miaoer Xu
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brian P Pollack
- Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA; Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Edwin M Horwitz
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lindsey Seldin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA; Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Chaoran Li
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
50
|
Abstract
Bone marrow is known as the site of hematopoiesis. What is not being described in textbooks of immunology is the fact that bone marrow is not only a generative, but also an antigen-responsive, immune organ. It is also a major storage site for antigen-specific memory B and T cells. That bone marrow is a priming site for T cell responses to blood borne antigens was discovered exactly 20 years ago. This review celebrates this important discovery. The review provides a number of examples of medical relevance of bone marrow as a central immune system, including cancer, microbial infections, autoimmune reactions, and bone marrow transplantation. Bone marrow mesenchymal stem cell-derived stromal cells provide distinct bone marrow niches for stem cells and immune cells. By transmitting anti-inflammatory dampening effects, facilitating wound healing and tissue regeneration mesenchymal stem cells contribute to homeostasis of bone and other tissues. Based on the evidence presented, the review proposes that bone marrow is a multifunctional and protective immune system. In an analogy to the central nervous system, it is suggested that bone marrow be designated as the central immune system.
Collapse
|