1
|
Liu Y, Liu Z, Li D, He X, Xiang L, Li B, Zhang C. Emerging role of regulatory T cells in the immunopathogenesis of vitiligo and implications for treatment. Br J Dermatol 2025; 192:796-806. [PMID: 39673777 DOI: 10.1093/bjd/ljae472] [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: 07/07/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/16/2024]
Abstract
Vitiligo is an autoimmune skin disease that targets pigment-producing melanocytes and results in depigmentation. This disfiguring condition frequently affects visible areas of the body and therefore causes a heavy psychological burden and a decreased quality of life. Although it remains intractable, the ever-growing understanding of its immunopathogenesis has dramatically shaped the treatment paradigm for vitiligo. With the impact of autoreactive cytotoxic T cells explained extensively, accumulating evidence suggests the unique role of regulatory T cells (Tregs) in the immune microenvironment of vitiligo. We systematically reviewed Treg deficiency, instability, reduced vitality and dysfunction in people with vitiligo, combined with novel findings regarding Treg function modulation in autoimmune backgrounds, including metabolic alteration, post-translational modifications and interaction with other immune cells. We further summarized classic and advanced Treg-targeted therapeutics in vitiligo practice and research. Herein, we share up-to-date knowledge of Tregs in vitiligo, providing insights into novel Treg-based therapeutic strategies.
Collapse
Affiliation(s)
- Yang Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ziqi Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai, China
| | - Xuanxuan He
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Leihong Xiang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai, China
| | - Chengfeng Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Rafiqi SI, Aldasouqi A, Paparodis RD, Dewan S, Farooqi A, Faisal S, Nemat Y, Salim N, Qureshi S, Mahmood A, Tovar Y, Jun JY, Kalinoski AL, Mirmira RG, Jaume JC, Imam S. Conversion of T Effector Cells Into T Regulatory Cells in Type 1 Diabetes/Latent Autoimmune Diabetes of Adults by Inhibiting eIF5A and Notch Pathways. Immunotargets Ther 2025; 14:205-226. [PMID: 40099149 PMCID: PMC11912933 DOI: 10.2147/itt.s504555] [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: 11/07/2024] [Accepted: 02/22/2025] [Indexed: 03/19/2025] Open
Abstract
Background The generation of functionally active, stable T regulatory cells (Tregs) is a crucial target of type 1 diabetes (T1D) immunotherapy. This study investigated therapeutic intervention for T1D/Latent autoimmune diabetes in adults (LADA), wherein the diabetogenic proinflammatory Treg (intermediate) cell subset was characterized and driven to a Treg phenotype (CD4+CD25+FOXP3+). This involved simultaneous inhibition of the eukaryotic initiation factor 5a (eIF5a) and Notch pathways using GC7 (N1-Guanyl-1,7-diaminoheptane) and Anti-DLL4 (Delta-like-ligand-4). Methods Peripheral blood from patients with T1D/LADA and healthy adults (n=7 each) was used to isolate the CD4+CD25- T cell population and CD4 deficient peripheral blood mononuclear cells (PBMCs). Cells were subjected to GAD65+GC7+anti-DLL4 treatment for seven days and compared with conventional anti-CD3/CD28/CD137 stimulation for conversion into the Tregs. Newly plasticized Tregs were assessed for their suppressive potential against freshly isolated autologous T responder cells. In addition, live, dead, and apoptotic cell counts were performed to evaluate the adverse effects of immunomodulatory treatment on immune cells. The data was analyzed with GraphPad Prism using 1- or 2-way ANOVA and a Student's t-test. Results A unique population of proinflammatory cytokines expressing intermediate Tregs (CD4+CD25-IFNg+IL17+FOXP3+) was characterized in T1D/LADA patients and found significantly increased compared to age-matched healthy adults. Simultaneous inhibition of eIF5a and Notch pathways could induce Treg phenotype in Treg-deficient CD4+ T cells and CD4 deficient PBMCs from T1D/LADA patients. GAD65+GC7+anti-DLL4 treatment plasticized Tregs withstanding a proinflammatory milieu mimicking T1D/LADA, and the plasticized Tregs exhibited a stable and suppressive functional phenotype. Furthermore, GAD65+GC7+anti-DLL4 treatment had no adverse effects on immune cells.The present approach is a multipronged approach involving the inhibition of eIF5a and Notch pathways that addresses the upregulation of immune tolerance, differentiation, and proliferation of cytotoxic T cells and alleviates β-cell dysfunction. Additionally, this treatment strategy could also be leveraged to boost Treg generation following islet transplantation or as a combinational therapy along with adoptive cell transfer.
Collapse
Affiliation(s)
- Shafiya Imtiaz Rafiqi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Ahmad Aldasouqi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH, USA
| | - Rodis D Paparodis
- Hellenic Endocrine Network, Athens, Greece, Endocrinology, Diabetes and Metabolism Clinics, Private Practice, Patras, Greece
- Stritch School of Medicine/Edward Hines, Jr. VA Hospital, Loyola University Chicago, Hines, IL, USA
| | - Sandesh Dewan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Aneeba Farooqi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Sarah Faisal
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- College of Art and Science, Case Western Reserve University, Cleveland, OH, USA
| | - Yousuf Nemat
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Florida Atlantic University, Boca Raton, FL, USA
| | - Nancy Salim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Salauddin Qureshi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, India
| | - Asif Mahmood
- University of Toledo Medical Centre, Hospital Medicine, University of Toledo, Toledo, OH, USA
| | - Yara Tovar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - John Y Jun
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Andrea L Kalinoski
- Department of Surgery, Integrated Core Facilities, University of Toledo, Toledo, OH, USA
| | | | - Juan Carlos Jaume
- Stritch School of Medicine/Edward Hines, Jr. VA Hospital, Loyola University Chicago, Hines, IL, USA
| | - Shahnawaz Imam
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
3
|
Zhou J, Felix FA, Jiang Y, Li D, Kim MC, Jang D, Cha S, Yu Q. Altered characteristics of regulatory T cells in target tissues of Sjögren's syndrome in murine models. Mol Immunol 2024; 174:47-56. [PMID: 39197397 PMCID: PMC11500054 DOI: 10.1016/j.molimm.2024.08.003] [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: 04/20/2024] [Revised: 07/31/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024]
Abstract
Sjӧgren's syndrome (SS), also known as Sjögren's disease, is a chronic autoimmune condition predominantly affecting the salivary and lacrimal glands. The disease is driven by autoimmune responses involving the activation and actions of major innate- and adaptive immune cell subsets. However, the specific characteristics and roles of regulatory T cells (Tregs) in SS remain elusive. This study seeks to clarify the main phenotypic and functional attributes of Tregs in the salivary glands and their draining lymph nodes in murine models of SS. Our flow cytometric analysis revealed that Tregs in the salivary gland-draining lymph nodes of female non-obese diabetic (NOD) mice, a spontaneous model of SS, exhibited a greater proportion of activated Tregs and fewer resting Tregs compared to Balb/c mice. Furthermore, Tregs from the salivary gland-draining lymph nodes of female C57BL/6.NOD-Aec1Aec2 (B6.NOD-Aec) mice, a model for primary SS, demonstrated significantly lower IL-10 production but markedly higher IFNγ- and IL-17 production than their C57BL/6 counterparts. Additionally, treatment of C57BL/6 Tregs with IL-7, a cytokine critical for SS pathogenesis, resulted in diminished IL-10 production and enhanced IFNγ and IL-17 production in these cells. Notably, the alterations in B6.NOD-Aec Tregs also included an increased expression of the immune-inhibitory molecule CTLA-4 compared to the C57BL/6 Tregs. Intriguingly, in vitro co-cultures of Tregs with conventional CD4 T cells and other key immune populations from lymph nodes indicated that Tregs from salivary gland-draining lymph nodes of both B6.NOD-Aec and C57BL/6 strains exhibited comparable and limited immunosuppressive effects on the proliferation and function of conventional CD4 T cells. The ability of B6.NOD-Aec Tregs to directly inflict damages to salivary gland epithelial tissues and contribute to SS pathologies through IFNγ and IL-17 that they produce warrants further investigations. In addition, enhancing the relatively weak immunosuppressive capacities of these Tregs may also serve as a viable strategy to alleviate the SS phenotype in the mouse models and potentially in patients.
Collapse
Affiliation(s)
- Jing Zhou
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA
| | - Fernanda Aragão Felix
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA; Department of Oral Surgery, Pathology, and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Yuqiao Jiang
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA
| | - Dongfang Li
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA
| | - Myung-Chul Kim
- Veterinary Diagnostic Laboratory Medicine, College of Veterinary Medicine, Jeju National University, 102, Jejudaehak-ro, Jeju-si, Jeju-do 63243, South Korea
| | - Daesong Jang
- Department of Oral & Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, FL 32610, USA; Center for Orphaned Autoimmune Disorders, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Seunghee Cha
- Department of Oral & Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, FL 32610, USA; Center for Orphaned Autoimmune Disorders, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Qing Yu
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA.
| |
Collapse
|
4
|
Contreras-Castillo E, García-Rasilla VY, García-Patiño MG, Licona-Limón P. Stability and plasticity of regulatory T cells in health and disease. J Leukoc Biol 2024; 116:33-53. [PMID: 38428948 DOI: 10.1093/jleuko/qiae049] [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: 11/14/2023] [Revised: 02/09/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024] Open
Abstract
The mechanisms that negatively regulate inflammation upon a pathogenic stimulus are crucial for the maintenance of tissue integrity and organ function. T regulatory cells are one of the main drivers in controlling inflammation. The ability of T regulatory cells to adapt to different inflammatory cues and suppress inflammation is one of the relevant features of T regulatory cells. During this process, T regulatory cells express different transcription factors associated with their counterparts, Th helper cells, including Tbx21, GATA-3, Bcl6, and Rorc. The acquisition of this transcription factor helps the T regulatory cells to suppress and migrate to the different inflamed tissues. Additionally, the T regulatory cells have different mechanisms that preserve stability while acquiring a particular T regulatory cell subtype. This review focuses on describing T regulatory cell subtypes and the mechanisms that maintain their identity in health and diseases.
Collapse
Affiliation(s)
- Eugenio Contreras-Castillo
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - Verónica Yutsil García-Rasilla
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - María Guadalupe García-Patiño
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, CU Coyoacán, México City 04510, Mexico
| |
Collapse
|
5
|
Bonnin E, Rodrigo Riestra M, Marziali F, Mena Osuna R, Denizeau J, Maurin M, Saez JJ, Jouve M, Bonté PE, Richer W, Nevo F, Lemoine S, Girard N, Lefevre M, Borcoman E, Vincent-Salomon A, Baulande S, Moreau HD, Sedlik C, Hivroz C, Lennon-Duménil AM, Tosello Boari J, Piaggio E. CD74 supports accumulation and function of regulatory T cells in tumors. Nat Commun 2024; 15:3749. [PMID: 38702311 PMCID: PMC11068745 DOI: 10.1038/s41467-024-47981-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
Regulatory T cells (Tregs) are plastic cells playing a pivotal role in the maintenance of immune homeostasis. Tregs actively adapt to the microenvironment where they reside; as a consequence, their molecular and functional profiles differ among tissues and pathologies. In tumors, the features acquired by Tregs remains poorly characterized. Here, we observe that human tumor-infiltrating Tregs selectively overexpress CD74, the MHC class II invariant chain. CD74 has been previously described as a regulator of antigen-presenting cell biology, however its function in Tregs remains unknown. CD74 genetic deletion in human primary Tregs reveals that CD74KO Tregs exhibit major defects in the organization of their actin cytoskeleton and intracellular organelles. Additionally, intratumoral CD74KO Tregs show a decreased activation, a drop in Foxp3 expression, a low accumulation in the tumor, and consistently, they are associated with accelerated tumor rejection in preclinical models in female mice. These observations are unique to tumor conditions as, at steady state, CD74KO-Treg phenotype, survival, and suppressive capacity are unaffected in vitro and in vivo. CD74 therefore emerges as a specific regulator of tumor-infiltrating Tregs and as a target to interfere with Treg anti-tumor activity.
Collapse
MESH Headings
- T-Lymphocytes, Regulatory/immunology
- Animals
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/immunology
- Histocompatibility Antigens Class II/metabolism
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/genetics
- Humans
- Female
- Mice
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Tumor Microenvironment/immunology
- Neoplasms/immunology
- Neoplasms/genetics
- Neoplasms/metabolism
- Neoplasms/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
Collapse
Affiliation(s)
- Elisa Bonnin
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Maria Rodrigo Riestra
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Federico Marziali
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Rafael Mena Osuna
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Jordan Denizeau
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Mathieu Maurin
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
| | - Juan Jose Saez
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
| | - Mabel Jouve
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
| | - Pierre-Emmanuel Bonté
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
| | - Wilfrid Richer
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | | | | | - Nicolas Girard
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
- Paris Saclay University, UVSQ, Versailles, France
- Institut du Thorax Curie Montsouris, Institut Curie, Paris, France
| | - Marine Lefevre
- Pathology Department, Institut Mutualiste Montsouris, Paris, France
| | - Edith Borcoman
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - Anne Vincent-Salomon
- Institut du Thorax Curie Montsouris, Institut Curie, Paris, France
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL Research University, Paris, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, PSL Research University, Institut Curie Research Center, Paris, France
| | - Helene D Moreau
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
| | - Christine Sedlik
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Claire Hivroz
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France
| | | | - Jimena Tosello Boari
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France.
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France.
| | - Eliane Piaggio
- INSERM U932 Immunity and Cancer, PSL University, Institut Curie Research Center, Paris, France.
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France.
- Egle Therapeutics, Paris, France.
| |
Collapse
|
6
|
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
|
7
|
Harjacek M. Role of regulatory T cells in pathogenesis and therapeutics of spondyloarthritis. REGULATORY T CELLS AND AUTOIMMUNE DISEASES 2024:165-196. [DOI: 10.1016/b978-0-443-13947-5.00042-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
8
|
Shao TY, Jiang TT, Stevens J, Russi AE, Troutman TD, Bernieh A, Pham G, Erickson JJ, Eshleman EM, Alenghat T, Jameson SC, Hogquist KA, Weaver CT, Haslam DB, Deshmukh H, Way SS. Kruppel-like factor 2+ CD4 T cells avert microbiota-induced intestinal inflammation. Cell Rep 2023; 42:113323. [PMID: 37889750 PMCID: PMC10822050 DOI: 10.1016/j.celrep.2023.113323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 09/05/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Intestinal colonization by antigenically foreign microbes necessitates expanded peripheral immune tolerance. Here we show commensal microbiota prime expansion of CD4 T cells unified by the Kruppel-like factor 2 (KLF2) transcriptional regulator and an essential role for KLF2+ CD4 cells in averting microbiota-driven intestinal inflammation. CD4 cells with commensal specificity in secondary lymphoid organs and intestinal tissues are enriched for KLF2 expression, and distinct from FOXP3+ regulatory T cells or other differentiation lineages. Mice with conditional KLF2 deficiency in T cells develop spontaneous rectal prolapse and intestinal inflammation, phenotypes overturned by eliminating microbiota or reconstituting with donor KLF2+ cells. Activated KLF2+ cells selectively produce IL-10, and eliminating IL-10 overrides their suppressive function in vitro and protection against intestinal inflammation in vivo. Together with reduced KLF2+ CD4 cell accumulation in Crohn's disease, a necessity for the KLF2+ subpopulation of T regulatory type 1 (Tr1) cells in sustaining commensal tolerance is demonstrated.
Collapse
Affiliation(s)
- Tzu-Yu Shao
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Tony T Jiang
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Joseph Stevens
- Division of Neonatology and Pulmonary Biology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Abigail E Russi
- Division of Gastroenterology, Hepatology and Advanced Nutrition, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Ty D Troutman
- Division of Allergy and Immunology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Anas Bernieh
- Division of Pathology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Giang Pham
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - John J Erickson
- Division of Neonatology and Pulmonary Biology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Emily M Eshleman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Theresa Alenghat
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Kristin A Hogquist
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Casey T Weaver
- Program in Immunology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35233, USA
| | - David B Haslam
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Hitesh Deshmukh
- Division of Neonatology and Pulmonary Biology, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
9
|
Zhang L, Shi X, Qiu H, Liu S, Yang T, Li X, Liu X. Protein modification by short-chain fatty acid metabolites in sepsis: a comprehensive review. Front Immunol 2023; 14:1171834. [PMID: 37869005 PMCID: PMC10587562 DOI: 10.3389/fimmu.2023.1171834] [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: 02/22/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Sepsis is a major life-threatening syndrome of organ dysfunction caused by a dysregulated host response due to infection. Dysregulated immunometabolism is fundamental to the onset of sepsis. Particularly, short-chain fatty acids (SCFAs) are gut microbes derived metabolites serving to drive the communication between gut microbes and the immune system, thereby exerting a profound influence on the pathophysiology of sepsis. Protein post-translational modifications (PTMs) have emerged as key players in shaping protein function, offering novel insights into the intricate connections between metabolism and phenotype regulation that characterize sepsis. Accumulating evidence from recent studies suggests that SCFAs can mediate various PTM-dependent mechanisms, modulating protein activity and influencing cellular signaling events in sepsis. This comprehensive review discusses the roles of SCFAs metabolism in sepsis associated inflammatory and immunosuppressive disorders while highlights recent advancements in SCFAs-mediated lysine acylation modifications, such as substrate supplement and enzyme regulation, which may provide new pharmacological targets for the treatment of sepsis.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Xinhui Shi
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Hongmei Qiu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Sijia Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Ting Yang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Xin Liu
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
10
|
Alvarez F, Piccirillo CA. The functional adaptation of effector Foxp3 + regulatory T cells to pulmonary inflammation. Eur J Immunol 2023; 53:e2250273. [PMID: 37366319 DOI: 10.1002/eji.202250273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023]
Abstract
During infections, the timings of effector differentiation of pulmonary immune responses are of paramount importance, as pathogen persistence and unsuppressed inflammation can rapidly lead to a loss of function, increased frailty, and death. Thus, both an efficient clearance of the danger and a rapid resolution of inflammation are critical to host survival. We now know that tissue-localized FoxP3+ regulatory T cells, a subset of CD4+ T cells, are highly attuned to the type of immune response, acquiring unique phenotypic characteristics that allow them to adapt their suppressive functions with the nature of inflammatory cells. To achieve this, activated effector TREG cells acquire specialized TH 1, TH 2, and TH 17-like characteristics that allow them to migrate, survive, and time their function(s) through refined mechanisms. Herein, we describe how this process requires a unique developmental path that includes the acquisition of master transcription factors and the expression of receptors adapted to sense local danger signals that are found during pulmonary inflammation. In turn, we offer an overview of how these characteristics promote the capacity of local effector TREG cells to proliferate, survive, and display suppressive strategies to resolve lung injury.
Collapse
Affiliation(s)
- Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
- Centre of Excellence in Translational Immunology (CETI), McGill University, Montréal, Québec, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
- Centre of Excellence in Translational Immunology (CETI), McGill University, Montréal, Québec, Canada
| |
Collapse
|
11
|
Okamoto M, Sasai M, Kuratani A, Okuzaki D, Arai M, Wing JB, Sakaguchi S, Yamamoto M. A genetic method specifically delineates Th1-type Treg cells and their roles in tumor immunity. Cell Rep 2023; 42:112813. [PMID: 37440410 DOI: 10.1016/j.celrep.2023.112813] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 04/06/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Regulatory T (Treg) cells expressing the transcription factor (TF) Foxp3 also express other TFs shared by T helper (Th) subsets under certain conditions. Here, to determine the roles of T-bet-expressing Treg cells, we generate a mouse strain, called VeDTR, in which T-bet/Foxp3 double-positive cells are engineered to be specifically labeled and depleted by a combination of Cre- and Flp-recombinase-dependent gene expression control. Characterization of T-bet+Foxp3+ cells using VeDTR mice reveals high resistance under oxidative stress, which is involved in accumulation of T-bet+Foxp3+ cells in tumor tissues. Moreover, short-term depletion of T-bet+Foxp3+ cells leads to anti-tumor immunity but not autoimmunity, whereas that of whole Treg cells does both. Although ablation of T-bet+Foxp3+ cells during Toxoplasma infection slightly enhances Th1 immune responses, it does not affect the course of the infection. Collectively, the intersectional genetic method reveals the specific roles of T-bet+Foxp3+ cells in suppressing tumor immunity.
Collapse
Affiliation(s)
- Masaaki Okamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan; Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan; Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan; Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ayumi Kuratani
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan; Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masaya Arai
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - James B Wing
- Laboratory of Human Immunology (Single Cell Immunology), WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan; Human Immunology Team, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan; Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan; Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
12
|
Zhu SY, Jiang YZ, Shen N, Li M, Yin HJ, Qiao JB. Changes in the intestinal microbiota of children with hand, foot, and mouth disease under 3 years old. Medicine (Baltimore) 2023; 102:e33687. [PMID: 37145009 PMCID: PMC10158917 DOI: 10.1097/md.0000000000033687] [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: 02/22/2023] [Accepted: 04/13/2023] [Indexed: 05/06/2023] Open
Abstract
This study aimed to clarify the characteristics of intestinal microbiota in children with hand, foot, and mouth disease (HFMD) under 3 years old. Fresh feces were collected from 54 children with HFMD and 30 healthy children. All of them were <3 years old. Sequencing of the 16S rDNA amplicons was performed. Between the 2 groups, the richness, diversity, and structure of the intestinal microbiota were analyzed by α-diversity and β-diversity. Linear discriminant analysis and LEfSe analyses were used to compare different bacterial classifications. The sex and age of the children in the 2 groups were not statistically significant (P = .92 and P = .98, respectively). Compared to healthy children, the Shannon index, Ace index, and Chao index were lower in children with HFMD (P = .027, P = .012, and P = .012, respectively). Based on the weighted or unweighted UniFrac distance analysis, the structure of the intestinal microbiota in HFMD was also significantly changed (P = .002 and P < .001, respectively). Linear discriminant analysis and LEfSe analysis showed that the changes of key bacteria were manifested as a decrease in Prevotella and Clostridium_XIVa (P < .001 and P < .001, respectively), while Escherichia and Bifidobacterium increased (P = .025 and P = .001, respectively). Children with HFMD under 3 years of age have intestinal microbiota disorder and show a decrease in diversity and richness. The decrease in the abundance of Prevotella and Clostridium, which can produce short-chain fatty acids, is also one of the characteristics of the change. These results can offer a theoretical foundation for the pathogenesis and microecological treatment of HFMD in infants.
Collapse
Affiliation(s)
- Su Yue Zhu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, China
| | - Ya Zhou Jiang
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, China
| | - Nan Shen
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, China
- Department of Hematology, The Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Min Li
- Department of Hematology, The Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Han Jun Yin
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, China
| | - Ji Bing Qiao
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, China
| |
Collapse
|
13
|
Malviya V, Yshii L, Junius S, Garg AD, Humblet-Baron S, Schlenner SM. Regulatory T-cell stability and functional plasticity in health and disease. Immunol Cell Biol 2023; 101:112-129. [PMID: 36479949 DOI: 10.1111/imcb.12613] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
FOXP3-expressing regulatory T cells (Treg ) are indispensable for immune homeostasis and tolerance, and in addition tissue-resident Treg have been found to perform noncanonical, tissue-specific functions. For optimal tolerogenic function during inflammatory disease, Treg are equipped with mechanisms that assure lineage stability. Treg lineage stability is closely linked to the installation and maintenance of a lineage-specific epigenetic landscape, specifically a Treg -specific DNA demethylation pattern. At the same time, for local and directed immune regulation Treg must possess a level of functional plasticity that requires them to partially acquire T helper cell (TH ) transcriptional programs-then referred to as TH -like Treg . Unleashing TH programs in Treg , however, is not without risk and may threaten the epigenetic stability of Treg with consequently pathogenic ex-Treg contributing to (auto-) inflammatory conditions. Here, we review how the Treg -stabilizing epigenetic landscape is installed and maintained, and further discuss the development, necessity and lineage instability risks of TH 1-, TH 2-, TH 17-like Treg and follicular Treg .
Collapse
Affiliation(s)
- Vanshika Malviya
- Department of Microbiology, Immunology and Transplantation, KU Leuven, University of Leuven, Leuven, Belgium
| | - Lidia Yshii
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Steffie Junius
- Department of Microbiology, Immunology and Transplantation, KU Leuven, University of Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stephanie Humblet-Baron
- Department of Microbiology, Immunology and Transplantation, KU Leuven, University of Leuven, Leuven, Belgium
| | - Susan M Schlenner
- Department of Microbiology, Immunology and Transplantation, KU Leuven, University of Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Liu X, Zhou M, Dai Z, Luo S, Shi Y, He Z, Chen Y. Salidroside alleviates ulcerative colitis via inhibiting macrophage pyroptosis and repairing the dysbacteriosis-associated Th17/Treg imbalance. Phytother Res 2023; 37:367-382. [PMID: 36331009 DOI: 10.1002/ptr.7636] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/03/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by flora disequilibrium and mucosal immunity disorder. Here, we report that salidroside effectively restricts experimental colitis from two aspects of intestinal macrophage pyroptosis and dysbacteriosis-derived colonic Th17/Treg imbalance. In innate immunity, the upregulated TREM1 and pyroptosis-related proteins in inflamed colons were inhibited by salidroside administration and further experiments in vitro showed that salidroside suppressed LPS/ATP-induced bone marrow-derived macrophages (BMDMs) pyroptosis evident by the decline of LDH and IL-1β release as well as the protein level of NLRP3, caspase-1, and GSDMD p30. Moreover, the TREM1 inhibitor weakened the effect of salidroside on BMDMs pyroptosis, whereas salidroside still could downregulate TREM1 when NLRP3 was inhibited. In adaptive immunity, salidroside improved the gut microflora diversity and Th17/Treg ratio in DSS-induced mice, especially promoting the abundance of Firmicutes. Clearance of the gut flora blocked the benefit of salidroside on colonic inflammation and Th17/Treg adaptive immunity, but transplanting salidroside-treated foecal bacterium into flora-depleted wild mice reproduced the resistance of salidroside to gut inflammation. Taken together, our data demonstrated that salidroside protected experimental colitis via skewing macrophage pyroptosis and Th17/Treg balance, indicating its potential effect on UC and other immune disorders.
Collapse
Affiliation(s)
- Xiaoman Liu
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Mingxia Zhou
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Zhenzhen Dai
- Shanghai Institute for Pediatric Research, Shanghai, China
| | - Shangjian Luo
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yingying Shi
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Zhenjuan He
- Department of Neonatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yingwei Chen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Mu J, Lei L, Zheng Y, Li D, Li J, Fu Y, Wang G, Liu Y. Comparative study of subcutaneous, intramuscular, and oral administration of bovine pathogenic Escherichia coli bacterial ghost vaccine in mice. Front Immunol 2022; 13:1008131. [DOI: 10.3389/fimmu.2022.1008131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022] Open
Abstract
Escherichia coli is one of the most common bacterial pathogens in cattle. Prophylactic vaccines are considered promising strategies with the potential to reduce the incidence of colibacillosis. Some studies suggested that bacterial ghosts may serve as a novel approach for preventing bacterial infections. However, the roles of administration route on vaccine immunogenicity and efficacy have not been investigated. In this study, the efficacy of vaccination via different immune routes in generating humoral and cellular immune response was compared through subcutaneous (SC), intramuscular (IM), and oral (O) administration in female BALB/c mice with bacterial ghosts prepared using wild type Escherichia coli isolates CE9, while phosphate buffer saline (PBS) and inactivated vaccines containing aluminum adjuvants (Killed) were used as control. Our results showed that the plasmid pBV220-E-aa-SNA containing E. coli was efficiently cleaved at 42°C with 94.8% positive ratio as assessed by colony counts. Transmission electron microscopy (TEM) confirmed bacteria retained intact surface structure while devoid of cytoplasmic component. We found that total IgG titers in killed, IM and SC groups showed significant increase on 7, 14, 21 and 28 days post-immunization. The IgA level of the IM group was higher than that of all other groups on the 28th day. Meanwhile, four experimental groups showed a significant difference in IgA levels compared with PBS control. In the IM group, an increase in the relative percentages of CD3+CD4+ T cells was accompanied by an increase in the relative percentages of splenic CD3+CD8+ T cells. In comparison with the inactivated vaccine, intramuscular CE9 ghosts immunization elicited higher levels of IL-1β, IL-2, IL-6 and IL-12. Subcutaneous and intramuscular immunizations were significantly associated with improved survival in comparison with oral route, traditional vaccine and the control. Pathologic assessment revealed that less severe tissue damage and inflammation were found in lung, kidney, and intestine of IM group compared with other groups. The results above demonstrate that immunization of Escherichia coli CE9 ghosts via intramuscular injection elicits a more robust antigen-specific immune response in mice to prevent the Escherichia coli infection.
Collapse
|
16
|
Cheung J, Zahorowska B, Suranyi M, Wong JKW, Diep J, Spicer ST, Verma ND, Hodgkinson SJ, Hall BM. CD4 +CD25 + T regulatory cells in renal transplantation. Front Immunol 2022; 13:1017683. [PMID: 36426347 PMCID: PMC9681496 DOI: 10.3389/fimmu.2022.1017683] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/13/2022] [Indexed: 09/14/2023] Open
Abstract
The immune response to an allograft activates lymphocytes with the capacity to cause rejection. Activation of CD4+CD25+Foxp3+T regulatory cells (Treg) can down-regulate allograft rejection and can induce immune tolerance to the allograft. Treg represent <10% of peripheral CD4+T cells and do not markedly increase in tolerant hosts. CD4+CD25+Foxp3+T cells include both resting and activated Treg that can be distinguished by several markers, many of which are also expressed by effector T cells. More detailed characterization of Treg to identify increased activated antigen-specific Treg may allow reduction of non-specific immunosuppression. Natural thymus derived resting Treg (tTreg) are CD4+CD25+Foxp3+T cells and only partially inhibit alloantigen presenting cell activation of effector cells. Cytokines produced by activated effector cells activate these tTreg to more potent alloantigen-activated Treg that may promote a state of operational tolerance. Activated Treg can be distinguished by several molecules they are induced to express, or whose expression they have suppressed. These include CD45RA/RO, cytokine receptors, chemokine receptors that alter pathways of migration and transcription factors, cytokines and suppression mediating molecules. As the total Treg population does not increase in operational tolerance, it is the activated Treg which may be the most informative to monitor. Here we review the methods used to monitor peripheral Treg, the effect of immunosuppressive regimens on Treg, and correlations with clinical outcomes such as graft survival and rejection. Experimental therapies involving ex vivo Treg expansion and administration in renal transplantation are not reviewed.
Collapse
Affiliation(s)
- Jason Cheung
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
| | | | - Michael Suranyi
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | | | - Jason Diep
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Stephen T. Spicer
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Nirupama D. Verma
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Suzanne J. Hodgkinson
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Bruce M. Hall
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| |
Collapse
|
17
|
Jia H, Chen J, Zhang X, Bi K, Zhou H, Liu T, Xu J, Diao H. IL-17A produced by invariant natural killer T cells and CD3 + CD56 + αGalcer-CD1d tetramer - T cells promote liver fibrosis in patients with primary biliary cholangitis. J Leukoc Biol 2022; 112:1079-1087. [PMID: 35730799 DOI: 10.1002/jlb.2a0622-586rrrr] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 06/03/2022] [Indexed: 12/24/2022] Open
Abstract
Primary biliary cholangitis (PBC) is characterized as interlobular bile duct injury and fibrosis, which results from the loss of tolerance to self-antigens. However, the exact pathologic mechanism leading to injury and fibrosis in PBC patients is not fully understood. Therefore, in this study, we examined the role of the T cell subsets in PBC patients and healthy controls (HCs). A higher number of invariant Natual killer T (iNKT) cells as well as CD3+ CD56+ αGalcer-CD1d tetramer- T cells were found in patients with PBC compared with HCs. Moreover, these 2 T subpopulations produced significantly higher levels of Interleukin (IL)-17A in PBC patients than those in in HCs, which has also been positively correlated with the disease severity. Furthermore, the level of IL-17A produced by these 2 subpopulations was increased after stimulation of the autoantibodies in patients with PBC. Also, the elevated IL-17A levels promoted the PBC-related fibrosis, thus presenting a change in frequencies and functions of these cell phenotypes in the deterioration of the duct damage-related fibrosis. This study clarified PBC patients' distinct T subpopulations characteristics, providing evidence-based diagnostic and therapies for these patients. The correlation between unclassical T subsets and IL-17A may provide a novel target for the immunotherapy of PBC.
Collapse
Affiliation(s)
- Hongyu Jia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianing Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xujun Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefan Bi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hetong Zhou
- Department of Mental Health, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Tianxing Liu
- Department of Biological Sciences, University of Toronto, Toronto, Canada
| | - Jia Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Park HJ, Lee SW, Park YH, Kim TC, Van Kaer L, Hong S. CD1d-independent NK1.1+ Treg cells are IL2-inducible Foxp3+ T cells co-expressing immunosuppressive and cytotoxic molecules. Front Immunol 2022; 13:951592. [PMID: 36177042 PMCID: PMC9513232 DOI: 10.3389/fimmu.2022.951592] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Regulatory T cells (Treg) play pivotal roles in maintaining self-tolerance and preventing immunological diseases such as allergy and autoimmunity through their immunosuppressive properties. Although Treg cells are heterogeneous populations with distinct suppressive functions, expression of natural killer (NK) cell receptors (NKR) by these cells remains incompletely explored. Here we identified that a small population of Foxp3+CD4+ Treg cells in mice expresses the NK1.1 NKR. Furthermore, we found that rare NK1.1+ subpopulations among CD4+ Treg cells develop normally in the spleen but not the thymus through CD1d-independent pathways. Compared with NK1.1- conventional Treg cells, these NK1.1+ Treg cells express elevated Treg cell phenotypic hallmarks, pro-inflammatory cytokines, and NK cell-related cytolytic mediators. Our results suggest that NK1.1+ Treg cells are phenotypically hybrid cells sharing functional properties of both NK and Treg cells. Interestingly, NK1.1+ Treg cells preferentially expanded in response to recombinant IL2 stimulation in vitro, consistent with their increased IL2Rαβ expression. Moreover, DO11.10 T cell receptor transgenic NK1.1+ Treg cells were expanded in an ovalbumin antigen-specific manner. In the context of lipopolysaccharide-induced systemic inflammation, NK1.1+ Treg cells downregulated immunosuppressive molecules but upregulated TNFα production, indicating their plastic adaptation towards a more pro-inflammatory rather than regulatory phenotype. Collectively, we propose that NK1.1+ Treg cells might play a unique role in controlling inflammatory immune responses such as infection and autoimmunity.
Collapse
Affiliation(s)
- Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, South Korea
| | - Sung Won Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, South Korea
| | - Yun Hoo Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, South Korea
| | - Tae-Cheol Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, South Korea
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, South Korea
- *Correspondence: Seokmann Hong,
| |
Collapse
|
19
|
Li DY, Chen L, Miao SY, Zhou M, Wu JH, Sun SW, Liu LL, Qi C, Xiong XZ. Inducible Costimulator-C-X-C Motif Chemokine Receptor 3 Signaling is Involved in Chronic Obstructive Pulmonary Disease Pathogenesis. Int J Chron Obstruct Pulmon Dis 2022; 17:1847-1861. [PMID: 35991707 PMCID: PMC9386059 DOI: 10.2147/copd.s371801] [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: 04/22/2022] [Accepted: 07/31/2022] [Indexed: 11/23/2022] Open
Abstract
Background The role of inducible costimulator (ICOS) signaling in chronic obstructive pulmonary disease (COPD) has not been fully elucidated. Methods We compared the percentages of ICOS+ T cells and ICOS+ regulatory T (Treg) cells in CD4+ T cells and CD4+CD25+FOXP3+ Tregs, respectively, in the peripheral blood of smokers with or without COPD to those in healthy controls. We further characterized their phenotypes using flow cytometry. To investigate the influence of ICOS signaling on C-X-C motif chemokine receptor 3 (CXCR3) expression in COPD, we evaluated the expression levels of ICOS and CXCR3 in vivo and in vitro. Results ICOS expression was elevated on peripheral CD4+ T cells and CD4+ Tregs of COPD patients, which positively correlated with the severity of lung function impairment in patients with stable COPD (SCOPD), but not in patients with acute exacerbation of COPD (AECOPD). ICOS+CD4+ Tregs in patients with SCOPD expressed higher levels of coinhibitors, programmed cell death protein 1 (PD-1) and T-cell immunoreceptor with Ig and ITIM domains (TIGIT), than ICOS−CD4+ Tregs, whereas ICOS+CD4+ T cells mostly exhibited a central memory (CD45RA−CCR7+) or effector memory (CD45RA−CCR7−) phenotype, ensuring their superior potential to respond potently and quickly to pathogen invasion. Furthermore, increased percentages of CXCR3+CD4+ T cells and CXCR3+CD4+ Tregs were observed in the peripheral blood of patients with SCOPD, and the expression level of CXCR3 was higher in ICOS+CD4+ T cells than in ICOS−CD4+ T cells. The percentage of CXCR3+CD4+ T cells was even higher in the bronchoalveolar lavage fluid than in matched peripheral blood in SCOPD group. Lastly, in vitro experiments showed that ICOS induced CXCR3 expression on CD4+ T cells. Conclusions ICOS signaling is upregulated in COPD, which induces CXCR3 expression. This may contribute to increased numbers of CXCR3+ Th1 cells in the lungs of patients with COPD, causing inflammation and tissue damage.
Collapse
Affiliation(s)
- Dan-Yang Li
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Long Chen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Shuai-Ying Miao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Department of Critical Care Medicine, General Hospital of Pingmei Shenma Medical Group, Pingdingshan, 467000, People's Republic of China
| | - Mei Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Jiang-Hua Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Sheng-Wen Sun
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Lan-Lan Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Chang Qi
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xian-Zhi Xiong
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| |
Collapse
|
20
|
Th1-like Treg in vitiligo: An incompetent regulator in immune tolerance. J Autoimmun 2022; 131:102859. [DOI: 10.1016/j.jaut.2022.102859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/21/2022]
|
21
|
Kaur H, Ghorai SM. Role of Cytokines as Immunomodulators. IMMUNOMODULATORS AND HUMAN HEALTH 2022:371-414. [DOI: 10.1007/978-981-16-6379-6_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Warunek J, Jin RM, Blair SJ, Garis M, Marzullo B, Wohlfert EA. Tbet Expression by Regulatory T Cells Is Needed to Protect against Th1-Mediated Immunopathology during Toxoplasma Infection in Mice. Immunohorizons 2021; 5:931-943. [PMID: 34893511 DOI: 10.4049/immunohorizons.2100080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 11/19/2022] Open
Abstract
Toxoplasma gondii infection has proven to be an ideal model to understand the delicate balance between protective immunity and immune-mediated pathology during infection. Lethal infection causes a collapse of T regulatory cells (Tregs) mediated by the loss of IL-2 and conversion of Tregs to IFN-γ-producing cells. Importantly, these Tregs highly express the Th1 transcription factor Tbet. To determine the role of Tbet in Tregs, we infected Tbx21f/f -Foxp3YFPCre and control Foxp3YFPCre mice with the type II strain of T. gondii, ME49. The majority of Tbx21f/f -Foxp3YFPCre mice succumbed to a nonlethal dose. Notably, parasite burden was reduced in Tbx21f/f -Foxp3YFPCre compared with Foxp3YFPCre control mice. We found that Tbx21f/f -Foxp3YFPCre mice have significantly higher serum levels of proinflammatory cytokines IFN-γ and TNF-α, suggestive of a heightened immune response. To test if CD4+ T cells were driving immunopathology, we treated Tbx21f/f -Foxp3YFPCre mice with anti-CD4-depleting Abs and partially rescued these mice. Broad-spectrum antibiotic treatment also improved survival, demonstrating a role for commensal flora in immunopathology in Tbx21f/f -Foxp3YFPCre mice. RNA sequencing analysis reinforced that Tbet regulates several key cellular pathways, including leukocyte activation, regulation of lymphocyte activation, and cell cycle progression, that help to maintain fitness in Tregs during Th1 responses. Taken together, our data show an important role for Tbet in Tregs in preventing lethal immunopathology during T. gondii infection, further highlighting the protective role of Treg plasticity in controlling immune responses to infection and the microbiota.
Collapse
Affiliation(s)
- Jordan Warunek
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| | - Richard M Jin
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| | - Sarah J Blair
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| | - Matthew Garis
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| | - Brandon Marzullo
- Genomics and Bioinformatics Core, State University of New York at Buffalo, Amherst, NY
| | - Elizabeth A Wohlfert
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| |
Collapse
|
23
|
Baysal H, De Pauw I, Zaryouh H, Peeters M, Vermorken JB, Lardon F, De Waele J, Wouters A. The Right Partner in Crime: Unlocking the Potential of the Anti-EGFR Antibody Cetuximab via Combination With Natural Killer Cell Chartering Immunotherapeutic Strategies. Front Immunol 2021; 12:737311. [PMID: 34557197 PMCID: PMC8453198 DOI: 10.3389/fimmu.2021.737311] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Cetuximab has an established role in the treatment of patients with recurrent/metastatic colorectal cancer and head and neck squamous cell cancer (HNSCC). However, the long-term effectiveness of cetuximab has been limited by the development of acquired resistance, leading to tumor relapse. By contrast, immunotherapies can elicit long-term tumor regression, but the overall response rates are much more limited. In addition to epidermal growth factor (EGFR) inhibition, cetuximab can activate natural killer (NK) cells to induce antibody-dependent cellular cytotoxicity (ADCC). In view of the above, there is an unmet need for the majority of patients that are treated with both monotherapy cetuximab and immunotherapy. Accumulated evidence from (pre-)clinical studies suggests that targeted therapies can have synergistic antitumor effects through combination with immunotherapy. However, further optimizations, aimed towards illuminating the multifaceted interplay, are required to avoid toxicity and to achieve better therapeutic effectiveness. The current review summarizes existing (pre-)clinical evidence to provide a rationale supporting the use of combined cetuximab and immunotherapy approaches in patients with different types of cancer.
Collapse
Affiliation(s)
- Hasan Baysal
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Ines De Pauw
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Hannah Zaryouh
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium.,Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Jan Baptist Vermorken
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium.,Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Jorrit De Waele
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - An Wouters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
24
|
Gellatly KJ, Strassner JP, Essien K, Refat MA, Murphy RL, Coffin-Schmitt A, Pandya AG, Tovar-Garza A, Frisoli ML, Fan X, Ding X, Kim EE, Abbas Z, McDonel P, Garber M, Harris JE. scRNA-seq of human vitiligo reveals complex networks of subclinical immune activation and a role for CCR5 in T reg function. Sci Transl Med 2021; 13:eabd8995. [PMID: 34516831 DOI: 10.1126/scitranslmed.abd8995] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kyle J Gellatly
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - James P Strassner
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Kingsley Essien
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Maggi Ahmed Refat
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Rachel L Murphy
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Anthony Coffin-Schmitt
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Amit G Pandya
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrea Tovar-Garza
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael L Frisoli
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Xueli Fan
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Xiaolan Ding
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Evangeline E Kim
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Zainab Abbas
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Patrick McDonel
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Manuel Garber
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - John E Harris
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
25
|
Regulatory T Cells and Inflammatory Mediators in Autoimmune Disease. J Invest Dermatol 2021; 142:774-780. [PMID: 34284898 DOI: 10.1016/j.jid.2021.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/22/2022]
Abstract
Regulatory T cells (Tregs) play a critical role in regulating tissue inflammation. Reduced Treg numbers and/or suppressive function contribute to autoimmune disease. Tregs can adopt the transcriptional programming of T helper (Th) type-1/2/17 cells to optimally suppress these subsets. Under specific conditions, these Th-like Tregs lose suppressive capacity and release proinflammatory cytokines to promote inflammation. This Treg plasticity depends on inflammatory mediators in the local environment. In this study, we review how cytokines impact Treg function and may contribute to autoimmune disease. A comprehensive understanding of Th-like Tregs may elucidate novel and more focused therapeutic approaches.
Collapse
|
26
|
Baeten P, Van Zeebroeck L, Kleinewietfeld M, Hellings N, Broux B. Improving the Efficacy of Regulatory T Cell Therapy. Clin Rev Allergy Immunol 2021; 62:363-381. [PMID: 34224053 PMCID: PMC8256646 DOI: 10.1007/s12016-021-08866-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2021] [Indexed: 12/11/2022]
Abstract
Autoimmunity is caused by an unbalanced immune system, giving rise to a variety of organ-specific to system disorders. Patients with autoimmune diseases are commonly treated with broad-acting immunomodulatory drugs, with the risk of severe side effects. Regulatory T cells (Tregs) have the inherent capacity to induce peripheral tolerance as well as tissue regeneration and are therefore a prime candidate to use as cell therapy in patients with autoimmune disorders. (Pre)clinical studies using Treg therapy have already established safety and feasibility, and some show clinical benefits. However, Tregs are known to be functionally impaired in autoimmune diseases. Therefore, ex vivo manipulation to boost and stably maintain their suppressive function is necessary when considering autologous transplantation. Similar to autoimmunity, severe coronavirus disease 2019 (COVID-19) is characterized by an exaggerated immune reaction and altered Treg responses. In light of this, Treg-based therapies are currently under investigation to treat severe COVID-19. This review provides a detailed overview of the current progress and clinical challenges of Treg therapy for autoimmune and hyperinflammatory diseases, with a focus on recent successes of ex vivo Treg manipulation.
Collapse
Affiliation(s)
- Paulien Baeten
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Lauren Van Zeebroeck
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium.,VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research (IRC), Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Markus Kleinewietfeld
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium.,VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research (IRC), Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Niels Hellings
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Bieke Broux
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium. .,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium. .,Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
27
|
da Luz JCDS, Antunes F, Clavijo-Salomon MA, Signori E, Tessarollo NG, Strauss BE. Clinical Applications and Immunological Aspects of Electroporation-Based Therapies. Vaccines (Basel) 2021; 9:727. [PMID: 34358144 PMCID: PMC8310106 DOI: 10.3390/vaccines9070727] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022] Open
Abstract
Reversible electropermeabilization (RE) is an ultrastructural phenomenon that transiently increases the permeability of the cell membrane upon application of electrical pulses. The technique was described in 1972 by Neumann and Rosenheck and is currently used in a variety of applications, from medicine to food processing. In oncology, RE is applied for the intracellular transport of chemotherapeutic drugs as well as the delivery of genetic material in gene therapies and vaccinations. This review summarizes the physical changes of the membrane, the particularities of bleomycin, and the immunological aspects involved in electrochemotherapy and gene electrotransfer, two important EP-based cancer therapies in human and veterinary oncology.
Collapse
Affiliation(s)
- Jean Carlos dos Santos da Luz
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, São Paulo 01246-000, Brazil; (J.C.d.S.d.L.); (F.A.); (N.G.T.)
| | - Fernanda Antunes
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, São Paulo 01246-000, Brazil; (J.C.d.S.d.L.); (F.A.); (N.G.T.)
| | | | - Emanuela Signori
- Institute of Translational Pharmacology, CNR, 00133 Rome, Italy;
| | - Nayara Gusmão Tessarollo
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, São Paulo 01246-000, Brazil; (J.C.d.S.d.L.); (F.A.); (N.G.T.)
| | - Bryan E. Strauss
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, São Paulo 01246-000, Brazil; (J.C.d.S.d.L.); (F.A.); (N.G.T.)
| |
Collapse
|
28
|
Angelicola S, Ruzzi F, Landuzzi L, Scalambra L, Gelsomino F, Ardizzoni A, Nanni P, Lollini PL, Palladini A. IFN-γ and CD38 in Hyperprogressive Cancer Development. Cancers (Basel) 2021; 13:309. [PMID: 33467713 PMCID: PMC7830527 DOI: 10.3390/cancers13020309] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) improve the survival of patients with multiple types of cancer. However, low response rates and atypical responses limit their success in clinical applications. The paradoxical acceleration of tumor growth after treatment, defined as hyperprogressive disease (HPD), is the most difficult problem facing clinicians and patients alike. The mechanisms that underlie hyperprogression (HP) are still unclear and controversial, although different factors are associated with the phenomenon. In this review, we propose two factors that have not yet been demonstrated to be directly associated with HP, but upon which it is important to focus attention. IFN-γ is a key cytokine in antitumor response and its levels increase during ICI therapy, whereas CD38 is an alternative immune checkpoint that is involved in immunosuppressive responses. As both factors are associated with resistance to ICI therapy, we have discussed their possible involvement in HPD with the conclusion that IFN-γ may contribute to HP onset through the activation of the inflammasome pathway, immunosuppressive enzyme IDO1 and activation-induced cell death (AICD) in effector T cells, while the role of CD38 in HP may be associated with the activation of adenosine receptors, hypoxia pathways and AICD-dependent T-cell depletion.
Collapse
Affiliation(s)
- Stefania Angelicola
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Lorena Landuzzi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Laura Scalambra
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Francesco Gelsomino
- Divisione di Oncologia Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.G.); (A.A.)
| | - Andrea Ardizzoni
- Divisione di Oncologia Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.G.); (A.A.)
| | - Patrizia Nanni
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Arianna Palladini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| |
Collapse
|
29
|
Maier I, Liu J, Ruegger PM, Deutschmann J, Patsch JM, Helbich TH, Borneman J, Schiestl RH. Intestinal bacterial indicator phylotypes associate with impaired DNA double-stranded break sensors but augmented skeletal bone micro-structure. Carcinogenesis 2020; 41:483-489. [PMID: 31840161 DOI: 10.1093/carcin/bgz204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 12/17/2022] Open
Abstract
Intestinal microbiota are considered a sensor for molecular pathways, which orchestrate energy balance, immune responses, and cell regeneration. We previously reported that microbiota restriction promoted higher levels of systemic radiation-induced genotoxicity, proliferative lymphocyte activation, and apoptotic polarization of metabolic pathways. Restricted intestinal microbiota (RM) that harbors increased abundance of Lactobacillus johnsonii (LBJ) has been investigated for bacterial communities that correlated radiation-induced genotoxicity. Indicator phylotypes were more abundant in RM mice and increased in prevalence after whole body irradiation in conventional microbiota (CM) mice, while none of the same ten most abundant phylotypes were different in abundance between CM mice before and after heavy ion irradiation. Muribaculum intestinale was detected highest in female small intestines in RM mice, which were lacking Ureaplasma felinum compared with males, and thus these bacteria could be contributing to the differential amounts of radiation-induced systemic genotoxicity between the CM and RM groups. Helicobacter rodentium and M.intestinale were found in colons in the radiation-resistant CM phenotype. While the expression of interferon-γ was elevated in the small intestine, and lower in blood in CM mice, high-linear energy transfer radiation reduced transforming growth factor-β with peripheral interleukin (IL)-17 in RM mice, particularly in females. We found that female RM mice showed improved micro-architectural bone structure and anti-inflammatory radiation response compared with CM mice at a delayed phase 6 weeks postexposure to particle radiation. However, microbiota restriction reduced inflammatory markers of tumor necrosis factor in marrow, when IL-17 was reduced by intraperitoneal injection of IL-17 neutralizing antibody.
Collapse
Affiliation(s)
- Irene Maier
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA
| | - Jared Liu
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA
| | - Paul M Ruegger
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Julia Deutschmann
- Department for Radiologic Technology, University of Applied Sciences Wiener Neustadt for Business and Engineering Ltd., Lower Austria, Austria
| | - Janina M Patsch
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel,Vienna, Austria
| | - Thomas H Helbich
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel,Vienna, Austria
| | - James Borneman
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Robert H Schiestl
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA.,Department of Pathology, University of California, Los Angeles, CA, USA
| |
Collapse
|
30
|
Prasad Singh N, Nagarkatti M, Nagarkatti P. From Suppressor T cells to Regulatory T cells: How the Journey That Began with the Discovery of the Toxic Effects of TCDD Led to Better Understanding of the Role of AhR in Immunoregulation. Int J Mol Sci 2020; 21:E7849. [PMID: 33105907 PMCID: PMC7660163 DOI: 10.3390/ijms21217849] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) was identified in the early 1970s as a receptor for the ubiquitous environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin), which is a member of halogenated aromatic hydrocarbons (HAHs). TCDD was found to be highly toxic to the immune system, causing thymic involution and suppression of a variety of T and B cell responses. The fact that environmental chemicals cause immunosuppression led to the emergence of a new field, immunotoxicology. While studies carried out in early 1980s demonstrated that TCDD induces suppressor T cells that attenuate the immune response to antigens, further studies on these cells were abandoned due to a lack of specific markers to identify such cells. Thus, it was not until 2001 when FoxP3 was identified as a master regulator of Regulatory T cells (Tregs) that the effect of AhR activation on immunoregulation was rekindled. The more recent research on AhR has led to the emergence of AhR as not only an environmental sensor but also as a key regulator of immune response, especially the differentiation of Tregs vs. Th17 cells, by a variety of endogenous, microbial, dietary, and environmental ligands. This review not only discusses how the role of AhR emerged from it being an environmental sensor to become a key immunoregulator, but also confers the identification of new AhR ligands, which are providing novel insights into the mechanisms of Treg vs. Th17 differentiation. Lastly, we discuss how AhR ligands can trigger epigenetic pathways, which may provide new opportunities to regulate inflammation and treat autoimmune diseases.
Collapse
Affiliation(s)
| | | | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29208, USA; (N.P.S.); (M.N.)
| |
Collapse
|
31
|
Li DY, Xiong XZ. ICOS + Tregs: A Functional Subset of Tregs in Immune Diseases. Front Immunol 2020; 11:2104. [PMID: 32983168 PMCID: PMC7485335 DOI: 10.3389/fimmu.2020.02104] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/04/2020] [Indexed: 01/02/2023] Open
Abstract
Recent studies have reported the pathological effect of ICOS+ T cells, but ICOS signals also widely participate in anti-inflammatory responses, particularly ICOS+ regulatory T (Treg) cells. The ICOS signaling pathway endows Tregs with increased generation, proliferation, and survival abilities. Furthermore, there is enough evidence to suggest a superior capacity of ICOS+ Tregs, which is partly attributable to IL-10 induced by ICOS, yet the associated mechanism needs further investigation. In this review, we discuss the complicated role of ICOS+ Tregs in several classical autoimmune diseases, allergic diseases, and cancers and investigate the related therapeutic applications in these diseases. Moreover, we identify ICOS as a potential biomarker for disease treatment and prognostic prediction. In addition, we believe that anti-ICOS/ICOSL monoclonal antibodies exhibit excellent clinical application potential. A thorough understanding of the effect of ICOS+ Tregs and the holistic role of ICOS toward the immune system will help to improve the therapeutic schedule of diseases.
Collapse
Affiliation(s)
- Dan-Yang Li
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Zhi Xiong
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Fung TH, Yang KY, Lui KO. An emerging role of regulatory T-cells in cardiovascular repair and regeneration. Theranostics 2020; 10:8924-8938. [PMID: 32802172 PMCID: PMC7415793 DOI: 10.7150/thno.47118] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence has demonstrated that immune cells play an important role in the regulation of tissue repair and regeneration. After injury, danger signals released by the damaged tissue trigger the initial pro-inflammatory phase essential for removing pathogens or cellular debris that is later replaced by the anti-inflammatory phase responsible for tissue healing. On the other hand, impaired immune regulation can lead to excessive scarring and fibrosis that could be detrimental for the restoration of organ function. Regulatory T-cells (Treg) have been revealed as the master regulator of the immune system that have both the immune and regenerative functions. In this review, we will summarize their immune role in the induction and maintenance of self-tolerance; as well as their regenerative role in directing tissue specific response for repair and regeneration. The latter is clearly demonstrated when Treg enhance the differentiation of stem or progenitor cells such as satellite cells to replace the damaged skeletal muscle, as well as the proliferation of parenchymal cells including neonatal cardiomyocytes for functional regeneration. Moreover, we will also discuss the reparative and regenerative role of Treg with a particular focus on blood vessels and cardiac tissues. Last but not least, we will describe the ongoing clinical trials with Treg in the treatment of autoimmune diseases that could give clinically relevant insights into the development of Treg therapy targeting tissue repair and regeneration.
Collapse
|
33
|
Interplay between Cytokine Circuitry and Transcriptional Regulation Shaping Helper T Cell Pathogenicity and Plasticity in Inflammatory Bowel Disease. Int J Mol Sci 2020; 21:ijms21093379. [PMID: 32403220 PMCID: PMC7247009 DOI: 10.3390/ijms21093379] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder manifested as Crohn’s disease (CD) and ulcerative colitis (UC) characterized by intestinal inflammation and involves a dysregulated immune response against commensal microbiota through the activation of CD4 T helper cells. T helper cell differentiation to effector or regulatory phenotypes is controlled by cytokine networks and transcriptional regulators. Distinct polarized T helper cells are able to alter their phenotypes to adapt to diverse and fluctuating physiological environments. T helper cells exhibit intrinsic instability and flexibility to express cytokines of other lineages or transdifferentiate from one T helper cell type to another in response to various perturbations from physiological cytokine milieu as a means of promoting local immunity in response to injury or ensure tissue homeostasis. Furthermore, functional plasticity and diversity of T helper cells are associated with pathogenicity and are critical for immune homeostasis and prevention of autoimmunity. In this review, we provide deeper insights into the combinatorial extrinsic and intrinsic signals that control plasticity and transdifferentiation of T helper cells and also highlight the potential of exploiting the genetic reprogramming plasticity of T helper cells in the treatment of IBD.
Collapse
|
34
|
Genetic Variation in Type 1 Diabetes Reconfigures the 3D Chromatin Organization of T Cells and Alters Gene Expression. Immunity 2020; 52:257-274.e11. [PMID: 32049053 DOI: 10.1016/j.immuni.2020.01.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/23/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
Genetics is a major determinant of susceptibility to autoimmune disorders. Here, we examined whether genome organization provides resilience or susceptibility to sequence variations, and how this would contribute to the molecular etiology of an autoimmune disease. We generated high-resolution maps of linear and 3D genome organization in thymocytes of NOD mice, a model of type 1 diabetes (T1D), and the diabetes-resistant C57BL/6 mice. Multi-enhancer interactions formed at genomic regions harboring genes with prominent roles in T cell development in both strains. However, diabetes risk-conferring loci coalesced enhancers and promoters in NOD, but not C57BL/6 thymocytes. 3D genome mapping of NODxC57BL/6 F1 thymocytes revealed that genomic misfolding in NOD mice is mediated in cis. Moreover, immune cells infiltrating the pancreas of humans with T1D exhibited increased expression of genes located on misfolded loci in mice. Thus, genetic variation leads to altered 3D chromatin architecture and associated changes in gene expression that may underlie autoimmune pathology.
Collapse
|
35
|
Hong JY, Li SS, Hu TY, Liu ZQ, Yu D, Yu HQ, Guan L, Wu GH, Zeng HT, Liu ZG, Yang PC. Frontline Science: TLR3 activation inhibits food allergy in mice by inducing IFN-γ + Foxp3 + regulatory T cells. J Leukoc Biol 2019; 106:1201-1209. [PMID: 30997942 DOI: 10.1002/jlb.3hi0918-348rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/12/2019] [Accepted: 04/02/2019] [Indexed: 01/14/2023] Open
Abstract
The pathologic feature of food allergy (FA) is the aberrant Th2-biased immune response in the intestine. Regulatory T cells (Tregs) play an important role in the suppression of aberrant immune response. The activities of the TLRs regulate multiple cell functions. This study aims to investigate the role of TLR3 activation in the regulation of Th2-biased immune response in the intestine by the generation of inducible Tregs (iTregs). In this study, polyinosinic polycytidylic acid (polyI:C) was used as an activator of TLR3. An FA mouse model was developed to establish the Th2-biased inflammation in the intestine. The effects of TLR3 activation on the generation of iTreg were tested in the culture and in mice. We observed that exposure to polyI:C induced IFN-γ+ Foxp3+ iTregs in mouse intestine and in the culture. The IFN-γ+ Foxp3+ iTregs showed immune suppressive functions. Exposure to polyI:C increased T-bet levels in CD4+ T cells. The T-bet formed a complex with GATA3 to dissociate Foxp3 from GATA3/Foxp3 complex in CD4+ T cells. The Foxp3 thus gained the opportunity to move to TGF-β promoter to generate iTregs. Administration with polyI:C prevented the development of FA and inhibited existing FA. In conclusion, activation of TLR3 induces IFN-γ+ Foxp3+ Tregs, which can prevent FA development and inhibit existing FA in mice.
Collapse
Affiliation(s)
- Jing-Yi Hong
- Department of Allergy, the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Shan-Shan Li
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Tian-Yong Hu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Zhi-Qiang Liu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
- Brain-Body Institute, McMaster University, Hamilton, Ontario, Canada
| | - Dian Yu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Hai-Qiong Yu
- Department of Allergy, the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Li Guan
- Department of Allergy, the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Gao-Hui Wu
- Department of Allergy, the Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Hao-Tao Zeng
- Longgang ENT Hospital & Shenzhen ENT Institute, Shenzhen, China
| | - Zhi-Gang Liu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Ping-Chang Yang
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
36
|
Di Giovangiulio M, Rizzo A, Franzè E, Caprioli F, Facciotti F, Onali S, Favale A, Stolfi C, Fehling HJ, Monteleone G, Fantini MC. Tbet Expression in Regulatory T Cells Is Required to Initiate Th1-Mediated Colitis. Front Immunol 2019; 10:2158. [PMID: 31572375 PMCID: PMC6749075 DOI: 10.3389/fimmu.2019.02158] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/28/2019] [Indexed: 12/22/2022] Open
Abstract
In normal conditions gut homeostasis is maintained by the suppressive activity of regulatory T cells (Tregs), characterized by the expression of the transcription factor FoxP3. In human inflammatory bowel disease, which is believed to be the consequence of the loss of tolerance toward antigens normally contained in the gut lumen, Tregs have been found to be increased and functionally active, thus pointing against their possible role in the pathogenesis of this immune-mediated disease. Though, in inflammatory conditions, Tregs have been shown to upregulate the T helper (Th) type 1-related transcription factor Tbet and to express the pro-inflammatory cytokine IFNγ, thus suggesting that at a certain point of the inflammatory process, Tregs might contribute to inflammation rather than suppress it. Starting from the observation that Tregs isolated from the lamina propria of active but not inactive IBD patients or uninflamed controls express Tbet and IFNγ, we investigated the functional role of Th1-like Tregs in the dextran sulfate model of colitis. As observed in human IBD, Th1-like Tregs were upregulated in the inflamed lamina propria of treated mice and the expression of Tbet and IFNγ in Tregs preceded the accumulation of conventional Th1 cells. By using a Treg-specific Tbet conditional knockout, we demonstrated that Tbet expression in Tregs is required for the development of colitis. Indeed, Tbet knockout mice developed milder colitis and showed an impaired Th1 immune response. In these mice not only the Tbet deficient Tregs but also the Tbet proficient conventional T cells showed reduced IFNγ expression. However, Tbet deficiency did not affect the Tregs suppressive capacity in vitro and in vivo in the adoptive transfer model of colitis. In conclusion here we show that Tbet expression by Tregs sustains the early phase of the Th1-mediated inflammatory response in the gut.
Collapse
Affiliation(s)
| | - Angelamaria Rizzo
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Eleonora Franzè
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, IRCCS Cà Granda Fundation, Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Onali
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Agnese Favale
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Massimo C Fantini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
37
|
Jing S, Lu J, Song J, Luo S, Zhou L, Quan C, Xi J, Zhao C. Effect of low-dose rituximab treatment on T- and B-cell lymphocyte imbalance in refractory myasthenia gravis. J Neuroimmunol 2019; 332:216-223. [DOI: 10.1016/j.jneuroim.2019.05.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/04/2019] [Accepted: 05/04/2019] [Indexed: 01/09/2023]
|
38
|
Hyun KH, Gil KC, Kim SG, Park SY, Hwang KW. Delphinidin Chloride and Its Hydrolytic Metabolite Gallic Acid Promote Differentiation of Regulatory T cells and Have an Anti-inflammatory Effect on the Allograft Model. J Food Sci 2019; 84:920-930. [PMID: 30977922 DOI: 10.1111/1750-3841.14490] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/21/2018] [Accepted: 02/07/2019] [Indexed: 01/10/2023]
Abstract
Regulatory T cells (Tregs) control the reactivity of other T cells to prevent excessive inflammatory responses. They also plays a role in preventing autoimmune diseases; but when they are overproduced, they decreased vital immunity, which can lead to invasion of external pathogens. Therefore, it is most important in preventing the development of immune diseases to maintain the homeostasis of these cells. Delphinidin chloride is an anthocyanidin and known to have anti-oxidant activities. However, its structure is very unstable and easily decomposed. One of these degradation products is gallic acid, which also has anti-oxidant effects. In this study, we examined the effect of these materials on Tregs in controlling immune response. It was found that these materials further promote differentiation into Tregs, and TGF-β and IL-2 related signals are involved in this process. Furthermore, it was verified that a variety of immunosuppressive proteins were secreted more, and the function of induced Tregs was also increased. Finally, in the allograft model, we could find a decrease in activated T cells when these materials were treated because they increased differentiation into Tregs. Therefore, these two materials are expected to become new candidates for the treatment of diseases caused by excessive activation of immune cells, such as autoimmune diseases. PRACTICAL APPLICATION: Delphinidin, a kind of anthocyanin rich in pigmented fruits, and its hydrolytic metabolite, gallic acid, are known to have antimicrobial and anti-oxidant properties. In this experiment, it was shown that delphinidin and gallic acid had an effect of increasing the differentiation of regulatory T cells, and the effect of suppressing the function of memory T cells was also observed. Due to these functions, delphinidin and gallic acid might have the potential to be used as immune suppressive agents in organ transplant and autoimmune disease patients or be a model for food development associated with the immune system.
Collapse
Affiliation(s)
- Ki Hyeob Hyun
- Host Defense Modulation Lab, College of Pharmacy Chung-Ang Univ., Heukseok-ro 84, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Ki Cheol Gil
- Host Defense Modulation Lab, College of Pharmacy Chung-Ang Univ., Heukseok-ro 84, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Sung Gun Kim
- Laboratory of Pharmacology, College of Pharmacy, Dankook Univ., Cheonan, 31116, Republic of Korea
| | - So-Young Park
- Laboratory of Pharmacology, College of Pharmacy, Dankook Univ., Cheonan, 31116, Republic of Korea
| | - Kwang Woo Hwang
- Host Defense Modulation Lab, College of Pharmacy Chung-Ang Univ., Heukseok-ro 84, Dongjak-gu, Seoul, 06974, Republic of Korea
| |
Collapse
|
39
|
Robertson SA, Green ES, Care AS, Moldenhauer LM, Prins JR, Hull ML, Barry SC, Dekker G. Therapeutic Potential of Regulatory T Cells in Preeclampsia-Opportunities and Challenges. Front Immunol 2019; 10:478. [PMID: 30984163 PMCID: PMC6448013 DOI: 10.3389/fimmu.2019.00478] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/21/2019] [Indexed: 12/26/2022] Open
Abstract
Inflammation is a central feature and is implicated as a causal factor in preeclampsia and other hypertensive disorders of pregnancy. Inflammatory mediators and leukocytes, which are elevated in peripheral blood and gestational tissues, contribute to the uterine vascular anomalies and compromised placental function that characterize particularly the severe, early onset form of disease. Regulatory T (Treg) cells are central mediators of pregnancy tolerance and direct other immune cells to counteract inflammation and promote robust placentation. Treg cells are commonly perturbed in preeclampsia, and there is evidence Treg cell insufficiency predates onset of symptoms. A causal role is implied by mouse studies showing sufficient numbers of functionally competent Treg cells must be present in the uterus from conception, to support maternal vascular adaptation and prevent later placental inflammatory pathology. Treg cells may therefore provide a tractable target for both preventative strategies and treatment interventions in preeclampsia. Steps to boost Treg cell activity require investigation and could be incorporated into pregnancy planning and preconception care. Pharmacological interventions developed to target Treg cells in autoimmune conditions warrant consideration for evaluation, utilizing rigorous clinical trial methodology, and ensuring safety is paramount. Emerging cell therapy tools involving in vitro Treg cell generation and/or expansion may in time become relevant. The success of preventative and therapeutic approaches will depend on resolving several challenges including developing informative diagnostic tests for Treg cell activity applicable before conception or during early pregnancy, selection of relevant patient subgroups, and identification of appropriate windows of gestation for intervention.
Collapse
Affiliation(s)
- Sarah A. Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Ella S. Green
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Alison S. Care
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Lachlan M. Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | | | - M. Louise Hull
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Women's and Children's Hospital, Adelaide, SA, Australia
| | - Simon C. Barry
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Gustaaf Dekker
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
40
|
Rocamora-Reverte L, Tuzlak S, von Raffay L, Tisch M, Fiegl H, Drach M, Reichardt HM, Villunger A, Tischner D, Wiegers GJ. Glucocorticoid Receptor-Deficient Foxp3 + Regulatory T Cells Fail to Control Experimental Inflammatory Bowel Disease. Front Immunol 2019; 10:472. [PMID: 30936873 PMCID: PMC6431616 DOI: 10.3389/fimmu.2019.00472] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/21/2019] [Indexed: 01/12/2023] Open
Abstract
Activation of the immune system increases systemic adrenal-derived glucocorticoid (GC) levels which downregulate the immune response as part of a negative feedback loop. While CD4+ T cells are essential target cells affected by GC, it is not known whether these hormones exert their major effects on CD4+ helper T cells, CD4+Foxp3+ regulatory T cells (Treg cells), or both. Here, we generated mice with a specific deletion of the glucocorticoid receptor (GR) in Foxp3+ Treg cells. Remarkably, while basal Treg cell characteristics and in vitro suppression capacity were unchanged, Treg cells lacking the GR did not prevent the induction of inflammatory bowel disease in an in vivo mouse model. Under inflammatory conditions, GR-deficient Treg cells acquired Th1-like characteristics and expressed IFN-gamma, but not IL-17, and failed to inhibit pro-inflammatory CD4+ T cell expansion in situ. These findings reveal that the GR is critical for Foxp3+ Treg cell function and suggest that endogenous GC prevent Treg cell plasticity toward a Th1-like Treg cell phenotype in experimental colitis. When equally active in humans, a rationale is provided to develop GC-mimicking therapeutic strategies which specifically target Foxp3+ Treg cells for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Lourdes Rocamora-Reverte
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Selma Tuzlak
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Laura von Raffay
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Marcel Tisch
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Heidi Fiegl
- Department of Obstetrics and Gynecology, Innsbruck University Hospital, Innsbruck, Austria
| | - Mathias Drach
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Denise Tischner
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - G Jan Wiegers
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
41
|
Pandiyan P, Bhaskaran N, Zou M, Schneider E, Jayaraman S, Huehn J. Microbiome Dependent Regulation of T regs and Th17 Cells in Mucosa. Front Immunol 2019; 10:426. [PMID: 30906299 PMCID: PMC6419713 DOI: 10.3389/fimmu.2019.00426] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/18/2019] [Indexed: 12/19/2022] Open
Abstract
Mammals co-exist with resident microbial ecosystem that is composed of an incredible number and diversity of bacteria, viruses and fungi. Owing to direct contact between resident microbes and mucosal surfaces, both parties are in continuous and complex interactions resulting in important functional consequences. These interactions govern immune homeostasis, host response to infection, vaccination and cancer, as well as predisposition to metabolic, inflammatory and neurological disorders. Here, we discuss recent studies on direct and indirect effects of resident microbiota on regulatory T cells (Tregs) and Th17 cells at the cellular and molecular level. We review mechanisms by which commensal microbes influence mucosa in the context of bioactive molecules derived from resident bacteria, immune senescence, chronic inflammation and cancer. Lastly, we discuss potential therapeutic applications of microbiota alterations and microbial derivatives, for improving resilience of mucosal immunity and combating immunopathology.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Natarajan Bhaskaran
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mangge Zou
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Elizabeth Schneider
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sangeetha Jayaraman
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
42
|
Lucca LE, Axisa PP, Singer ER, Nolan NM, Dominguez-Villar M, Hafler DA. TIGIT signaling restores suppressor function of Th1 Tregs. JCI Insight 2019; 4:124427. [PMID: 30728325 DOI: 10.1172/jci.insight.124427] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/03/2019] [Indexed: 12/28/2022] Open
Abstract
Th1 Tregs are characterized by the acquisition of proinflammatory cytokine secretion and reduced suppressor activity. Th1 Tregs are found at increased frequency in autoimmune diseases, including type 1 diabetes and multiple sclerosis (MS). We have previously reported that in vitro stimulation with IL-12 recapitulates the functional and molecular features of MS-associated Th1 Tregs, revealing a central role for hyperactivation of the Akt pathway in their induction. TIGIT is a newly identified coinhibitory receptor that marks Tregs that specifically control Th1 and Th17 responses. Here, we report that signaling through TIGIT counteracts the action of IL-12 in inducing the Th1 program. Specifically, TIGIT signaling represses production of IFN-γ and T-bet expression and restores suppressor function in Tregs treated with IL-12. FoxO1 functional inhibition abolishes the protective effect of TIGIT, indicating that TIGIT signaling promotes FoxO1 nuclear localization. Consistent with this observation, signaling through TIGIT leads to a rapid suppression of Akt function and FoxO1 phosphorylation. Finally, TIGIT stimulation reduces the production of IFN-γ and corrects the suppressor defect of Tregs from patients with MS. Our results indicate an important role for TIGIT in controlling the functional stability of Tregs through repression of Akt, suggesting that the TIGIT pathway could be targeted for immunomodulatory therapies in human autoimmune disorders.
Collapse
Affiliation(s)
- Liliana E Lucca
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Pierre-Paul Axisa
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Emily R Singer
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Neal M Nolan
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
43
|
Kitz A, Singer E, Hafler D. Regulatory T Cells: From Discovery to Autoimmunity. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a029041. [PMID: 29311129 DOI: 10.1101/cshperspect.a029041] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is a genetically mediated autoimmune disease of the central nervous system. Allelic variants lead to lower thresholds of T-cell activation resulting in activation of autoreactive T cells. Environmental factors, including, among others, diet, vitamin D, and smoking, in combination with genetic predispositions, play a substantial role in disease development and activation of autoreactive T cells. FoxP3+ regulatory T cells (Tregs) have emerged as central in the control of autoreactive T cells. A consistent finding in patients with MS is defects in Treg cell function with reduced suppression of effector T cells and production of proinflammatory cytokines. Emerging data suggests that functional Tregs become effector-like T cells with loss of function associated with T-bet expression and interferon γ (IFN-γ) secretion.
Collapse
Affiliation(s)
- Alexandra Kitz
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| | - Emily Singer
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| | - David Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
44
|
Sorini C, Cardoso RF, Gagliani N, Villablanca EJ. Commensal Bacteria-Specific CD4 + T Cell Responses in Health and Disease. Front Immunol 2018; 9:2667. [PMID: 30524431 PMCID: PMC6256970 DOI: 10.3389/fimmu.2018.02667] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Over the course of evolution, mammalian body surfaces have adapted their complex immune system to allow a harmless coexistence with the commensal microbiota. The adaptive immune response, in particular CD4+ T cell-mediated, is crucial to maintain intestinal immune homeostasis by discriminating between harmless (e.g., dietary compounds and intestinal microbes) and harmful stimuli (e.g., pathogens). To tolerate food molecules and microbial components, CD4+ T cells establish a finely tuned crosstalk with the environment whereas breakdown of these mechanisms might lead to chronic disease associated with mucosal barriers and beyond. How commensal-specific immune responses are regulated and how these molecular and cellular mechanisms can be manipulated to treat chronic disorders is yet poorly understood. In this review, we discuss current knowledge of the regulation of commensal bacteria-specific CD4+ T cells. We place particular focus on the key role of commensal-specific CD4+ T cells in maintaining tolerance while efficiently eradicating local and systemic infections, with a focus on factors that trigger their aberrant activation.
Collapse
Affiliation(s)
- Chiara Sorini
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Rebeca F. Cardoso
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Nicola Gagliani
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eduardo J. Villablanca
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
45
|
Zhou G, Wu W, Yu L, Yu T, Yang W, Wang P, Zhang X, Cong Y, Liu Z. Tripartite motif-containing (TRIM) 21 negatively regulates intestinal mucosal inflammation through inhibiting TH1/TH17 cell differentiation in patients with inflammatory bowel diseases. J Allergy Clin Immunol 2018; 142:1218-1228.e12. [DOI: 10.1016/j.jaci.2017.09.038] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/26/2017] [Accepted: 09/12/2017] [Indexed: 01/18/2023]
|
46
|
Microbiota-derived short-chain fatty acids promote Th1 cell IL-10 production to maintain intestinal homeostasis. Nat Commun 2018; 9:3555. [PMID: 30177845 PMCID: PMC6120873 DOI: 10.1038/s41467-018-05901-2] [Citation(s) in RCA: 419] [Impact Index Per Article: 59.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/20/2018] [Indexed: 02/07/2023] Open
Abstract
T-cells are crucial in maintanence of intestinal homeostasis, however, it is still unclear how microbiota metabolites regulate T-effector cells. Here we show gut microbiota-derived short-chain fatty acids (SCFAs) promote microbiota antigen-specific Th1 cell IL-10 production, mediated by G-protein coupled receptors 43 (GPR43). Microbiota antigen-specific Gpr43−/− CBir1 transgenic (Tg) Th1 cells, specific for microbiota antigen CBir1 flagellin, induce more severe colitis compared with wide type (WT) CBir1 Tg Th1 cells in Rag−/− recipient mice. Treatment with SCFAs limits colitis induction by promoting IL-10 production, and administration of anti-IL-10R antibody promotes colitis development. Mechanistically, SCFAs activate Th1 cell STAT3 and mTOR, and consequently upregulate transcription factor B lymphocyte-induced maturation protein 1 (Blimp-1), which mediates SCFA-induction of IL-10. SCFA-treated Blimp1−/− Th1 cells produce less IL-10 and induce more severe colitis compared to SCFA-treated WT Th1 cells. Our studies, thus, provide insight into how microbiota metabolites regulate Th1 cell functions to maintain intestinal homeostasis. T cells play a critical role in intestinal homeostasis, with increasing evidence suggesting a role for the microbiome metabolome in modulating this response. Here the authors show short-chain fatty acids promote IL-10 production in Th1 cells.
Collapse
|
47
|
Affiliation(s)
- Tin Kyaw
- From the BakerIDI Heart and Diabetes Institute (T.K., B.-H.T., A.B.), Department of Immunology (A.B.); and Centre for Inflammatory Disorders (T.K., B.-H.T., A.B.), Monash University, Melbourne, Australia
| | - Ban-Hock Toh
- From the BakerIDI Heart and Diabetes Institute (T.K., B.-H.T., A.B.), Department of Immunology (A.B.); and Centre for Inflammatory Disorders (T.K., B.-H.T., A.B.), Monash University, Melbourne, Australia
| | - Alex Bobik
- From the BakerIDI Heart and Diabetes Institute (T.K., B.-H.T., A.B.), Department of Immunology (A.B.); and Centre for Inflammatory Disorders (T.K., B.-H.T., A.B.), Monash University, Melbourne, Australia.
| |
Collapse
|
48
|
Zhou G, Yu L, Fang L, Yang W, Yu T, Miao Y, Chen M, Wu K, Chen F, Cong Y, Liu Z. CD177 + neutrophils as functionally activated neutrophils negatively regulate IBD. Gut 2018; 67:1052-1063. [PMID: 28468761 DOI: 10.1136/gutjnl-2016-313535] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/20/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Neutrophils are accumulated in inflamed mucosa of IBD and play an important role in the pathogenesis. CD177 is expressed in neutrophils specifically and upregulated during inflammation. However, the role of CD177+ neutrophils in pathogenesis of IBD remains elusive. MATERIALS AND METHODS Expression of CD177 was analysed in peripheral blood and intestinal mucosa from patients with IBD using quantitative RT-PCR, flow cytometry and immunohistochemistry. CD177+ and CD177- neutrophils were isolated to determine gene differences by RNA sequencing. Colitis was established in CD177-/- and wild-type mice in response to dextran sulfate sodium (DSS) insults to determine the role of CD177+ neutrophils in IBD. RESULTS CD177+ neutrophils were markedly increased in peripheral blood and inflamed mucosa from patients with active IBD compared with healthy controls. RNA sequencing revealed that differential gene expression between CD177+ and CD177- neutrophils from patients with IBD was associated with response to bacterial defence, hydrogen peroxide and reactive oxygen species (ROS). CD177+ neutrophils produced lower levels of proinflammatory cytokines (ie, interferon-γ, interleukin (IL)-6, IL-17A), but higher levels of IL-22 and transforming growth factor-β, and exhibited increased bactericidal activities (ie, ROS, antimicrobial peptides, neutrophil extracellular trap) compared with CD177- subset. CD177-/- mice developed more severe colitis on DSS insults compared with wild-type mice. Moreover, CD177 deficiency led to compromised intestinal barrier and impaired antibacterial immunity through decreased production of IL-22 by CD177- neutrophils. CONCLUSIONS CD177+ neutrophils represent functionally activated population and play a protective role in IBD through increased bactericidal activity and IL-22 production. Targeting CD177+ neutrophils may be beneficial for treatment of IBD.
Collapse
Affiliation(s)
- Guangxi Zhou
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Lin Yu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Leilei Fang
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Wenjing Yang
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Tianming Yu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yinglei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kaichun Wu
- Department of Gastroenterology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Feidi Chen
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Zhanju Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
49
|
Castro F, Cardoso AP, Gonçalves RM, Serre K, Oliveira MJ. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front Immunol 2018; 9:847. [PMID: 29780381 PMCID: PMC5945880 DOI: 10.3389/fimmu.2018.00847] [Citation(s) in RCA: 855] [Impact Index Per Article: 122.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/05/2018] [Indexed: 12/15/2022] Open
Abstract
Interferon-gamma (IFN-γ) is a pleiotropic molecule with associated antiproliferative, pro-apoptotic and antitumor mechanisms. This effector cytokine, often considered as a major effector of immunity, has been used in the treatment of several diseases, despite its adverse effects. Although broad evidence implicating IFN-γ in tumor immune surveillance, IFN-γ-based therapies undergoing clinical trials have been of limited success. In fact, recent reports suggested that it may also play a protumorigenic role, namely, through IFN-γ signaling insensitivity, downregulation of major histocompatibility complexes, and upregulation of indoleamine 2,3-dioxygenase and of checkpoint inhibitors, as programmed cell-death ligand 1. However, the IFN-γ-mediated responses are still positively associated with patient's survival in several cancers. Consequently, major research efforts are required to understand the immune contexture in which IFN-γ induces its intricate and highly regulated effects in the tumor microenvironment. This review discusses the current knowledge on the pro- and antitumorigenic effects of IFN-γ as part of the complex immune response to cancer, highlighting the relevance to identify IFN-γ responsive patients for the improvement of therapies that exploit associated signaling pathways.
Collapse
Affiliation(s)
- Flávia Castro
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Patrícia Cardoso
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Raquel Madeira Gonçalves
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Karine Serre
- IMM – Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria José Oliveira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Departamento de Patologia e Oncologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
50
|
Grinberg-Bleyer Y, Caron R, Seeley JJ, De Silva NS, Schindler CW, Hayden MS, Klein U, Ghosh S. The Alternative NF-κB Pathway in Regulatory T Cell Homeostasis and Suppressive Function. THE JOURNAL OF IMMUNOLOGY 2018; 200:2362-2371. [PMID: 29459403 DOI: 10.4049/jimmunol.1800042] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022]
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) are essential regulators of immune responses. Perturbation of Treg homeostasis or function can lead to uncontrolled inflammation and autoimmunity. Therefore, understanding the molecular mechanisms involved in Treg biology remains an active area of investigation. It has been shown previously that the NF-κB family of transcription factors, in particular, the canonical pathway subunits, c-Rel and p65, are crucial for the development, maintenance, and function of Tregs. However, the role of the alternative NF-κB pathway components, p100 and RelB, in Treg biology remains unclear. In this article, we show that conditional deletion of the p100 gene, nfkb2, in Tregs, resulted in massive inflammation because of impaired suppressive function of nfkb2-deficient Tregs. Surprisingly, mice lacking RelB in Tregs did not exhibit the same phenotype. Instead, deletion of both relb and nfkb2 rescued the inflammatory phenotype, demonstrating an essential role for p100 as an inhibitor of RelB in Tregs. Our data therefore illustrate a new role for the alternative NF-κB signaling pathway in Tregs that has implications for the understanding of molecular pathways driving tolerance and immunity.
Collapse
Affiliation(s)
- Yenkel Grinberg-Bleyer
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Rachel Caron
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - John J Seeley
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Nilushi S De Silva
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Christian W Schindler
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Matthew S Hayden
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Ulf Klein
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032;
| |
Collapse
|