1
|
Accogli T, Hibos C, Milian L, Geindreau M, Richard C, Humblin E, Mary R, Chevrier S, Jacquin E, Bernard A, Chalmin F, Paul C, Ryffel B, Apetoh L, Boidot R, Bruchard M, Ghiringhelli F, Vegran F. The intrinsic expression of NLRP3 in Th17 cells promotes their protumor activity and conversion into Tregs. Cell Mol Immunol 2025; 22:541-556. [PMID: 40195474 PMCID: PMC12041534 DOI: 10.1038/s41423-025-01281-y] [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: 06/19/2024] [Accepted: 03/14/2025] [Indexed: 04/09/2025] Open
Abstract
Th17 cells can perform either regulatory or inflammatory functions depending on the cytokine microenvironment. These plastic cells can transdifferentiate into Tregs during inflammation resolution, in allogenic heart transplantation models, or in cancer through mechanisms that remain poorly understood. Here, we demonstrated that NLRP3 expression in Th17 cells is essential for maintaining their immunosuppressive functions through an inflammasome-independent mechanism. In the absence of NLRP3, Th17 cells produce more inflammatory cytokines (IFNγ, Granzyme B, TNFα) and exhibit reduced immunosuppressive activity toward CD8+ cells. Moreover, the capacity of NLRP3-deficient Th17 cells to transdifferentiate into Treg-like cells is lost. Mechanistically, NLRP3 in Th17 cells interacts with the TGF-β receptor, enabling SMAD3 phosphorylation and thereby facilitating the acquisition of immunosuppressive functions. Consequently, the absence of NLRP3 expression in Th17 cells from tumor-bearing mice enhances CD8 + T-cell effectiveness, ultimately inhibiting tumor growth.
Collapse
Affiliation(s)
- Théo Accogli
- INSERM, Dijon, France
- University of Burgundy, Dijon, France
| | | | - Lylou Milian
- INSERM, Dijon, France
- University of Burgundy, Dijon, France
- Unité de Biologie Moléculaire-Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | | | - Corentin Richard
- Unité de Biologie Moléculaire-Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | | | | | - Sandy Chevrier
- Unité de Biologie Moléculaire-Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Elise Jacquin
- INSERM, Dijon, France
- University of Burgundy, Dijon, France
| | | | - Fanny Chalmin
- Cancer Biology Transfer Platform, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Catherine Paul
- LIIC, EA7269, Université de Bourgogne Franche Comté, Dijon, France
- Immunology and Immunotherapy of Cancer Laboratory, EPHE, PSL Research University, Paris, France
| | - Berhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orléans, France
| | - Lionel Apetoh
- Brown Center for Immunotherapy, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Romain Boidot
- Unité de Biologie Moléculaire-Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | | | - François Ghiringhelli
- INSERM, Dijon, France
- University of Burgundy, Dijon, France
- Cancer Biology Transfer Platform, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
- Genetic and Immunology Medical Institute, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Frédérique Vegran
- INSERM, Dijon, France.
- University of Burgundy, Dijon, France.
- Unité de Biologie Moléculaire-Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France.
- Cancer Biology Transfer Platform, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France.
| |
Collapse
|
2
|
Tan Y, Chen X, Shen Y, Yang J, Wang B, Wang Y, Zhou W, Han Q, Yao Z, Li H, Liang J. Exploring T H17-mediated inflammation in epidermolytic ichthyosis: Clinical and mechanistic insight. Clin Immunol 2025; 277:110494. [PMID: 40288548 DOI: 10.1016/j.clim.2025.110494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/19/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025]
Abstract
Epidermolytic ichthyosis (EI) is a genetic skin disorder caused by mutations in the KRT1 and KRT10 genes, leading to severe skin abnormalities and inflammation. Current treatment options are limited, emphasizing the need for pathogenesis-based therapies. This study investigates the role of T helper type 17 (TH17) inflammatory responses in EI and explores the mechanisms underlying these responses. We found that patients from the family carrying both KRT1 and MPO mutations exhibited varying degrees of psoriasis-like manifestations and significant therapeutic responses to anti-IL-17 A treatment. The treatment efficacy was also confirmed in patients with KRT10 mutations. Mechanistically, Single-nucleus RNA sequencing revealed significantly elevated levels of TH-17 related cytokines in epidermis and CCR6+ TH17 cell infiltration in the dermis. Additionally, we observed aberrant activation of the IκBα-JNK-c-Jun signaling pathway, leading to heightened IL-8 expression and exacerbated inflammation. Our findings underscore the critical role of TH17-mediated inflammation in the pathogenesis of EI and suggest potential therapeutic avenues targeting this pathway to improve patient outcomes.
Collapse
Affiliation(s)
- Yidong Tan
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuanyi Chen
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yihang Shen
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinxiang Yang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bing Wang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yumeng Wang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weinan Zhou
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiuyi Han
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Yao
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Huaguo Li
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Jianying Liang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China; Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Nakashima M, Fukumoto A, Matsuda S. Beneficial Probiotics with New Cancer Therapies for Improved Treatment of Hepatocellular Carcinoma. Diseases 2025; 13:111. [PMID: 40277821 PMCID: PMC12025462 DOI: 10.3390/diseases13040111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant form of primary liver cancer. Intricate networks linked to the host immune system may be associated with the pathogenesis of HCC. A huge amount of interdisciplinary medical information for the treatment of HCC has been accumulated over recent years. For example, advances in new immunotherapy have improved the results of treatment for HCC. This approach can be advantageously combined with standard conventional treatments such as surgical resection to improve the therapeutic effect. However, several toxic effects of treatments may pose a significant threat to human health. Now, a shift in mindset is important for achieving superior cancer therapy, where probiotic therapy may be considered, at least within the bounds of safety. The interplay between the gut microbiota and immune system could affect the efficacy of several anticancer treatments, including of immune checkpoint therapy via the alteration of Th17 cell function against various malignant tumors. Here, some recent anticancer techniques are discussed, whereby the growth of HCC may be effectively and safely repressed by probiotic therapy.
Collapse
Affiliation(s)
| | | | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
4
|
Heredia M, Barendregt DMH, Tindemans I, Klomberg RCW, Aardoom MA, Calado B, Costes LMM, Joosse ME, Hulleman-van Haaften DH, Tuk B, van Berkel LA, Kemos P, Ruemmele FM, Croft NM, Escher JC, de Ridder L, Samsom JN. Gut-homing and intestinal TIGIT negCD38 + memory T cells acquire an IL-12-induced, ex-Th17 pathogenic phenotype in a subgroup of Crohn's disease patients with a severe disease course. Mucosal Immunol 2025; 18:299-311. [PMID: 39586377 DOI: 10.1016/j.mucimm.2024.11.008] [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: 06/01/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
CD4+ memory T cell (TM) reactivation drives chronicity in inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis. Defects driving loss of TM regulation likely differ between patients but remain undefined. In health, approximately 40 % of circulating gut-homing CD38+TM express co-inhibitory receptor T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT). TIGIT+CD38+TM have regulatory function while TIGITnegCD38+TM are enriched in IFN-γ-producing cells. We hypothesized TIGITnegCD38+TM are inflammatory and drive disease in a subgroup of IBD patients. We characterized TIGIT+CD38+TM in a uniquely large cohort of pediatric IBD patients from time of diagnosis into adulthood. Circulating TIGITnegCD38+TM frequencies were higher in a subgroup of therapy-naïve CD patients with high plasma IFN-γ and a more severe disease course. TIGITnegCD38+TM were highly enriched in HLA-DR+ and ex-Th17/Th1-like cells, high producers of IFN-γ. Cultures of healthy-adult-stimulated TM identified IL-12 as the only IBD-related inflammatory cytokine to drive the pathogenic ex-Th17-TIGITnegCD38+ phenotype. Moreover, IL12RB2 mRNA expression was higher in TIGITnegCD38+TM than TIGIT+CD38+TM, elevated in CD biopsies compared to controls, and correlated with severity of intestinal inflammation. Overall, we argue that in a subgroup of pediatric CD, increased IL-12 signaling drives reprogramming of Th17 to inflammatory Th1-like TIGITnegCD38+TM and causes more severe disease.
Collapse
Affiliation(s)
- Maud Heredia
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Daniëlle M H Barendregt
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Irma Tindemans
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Renz C W Klomberg
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Martine A Aardoom
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Beatriz Calado
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Léa M M Costes
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Maria E Joosse
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Daniëlle H Hulleman-van Haaften
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Bastiaan Tuk
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Lisette A van Berkel
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Polychronis Kemos
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Frank M Ruemmele
- Department of Pediatric Gastroenterology, Necker-Enfants Malades University Hospital, Institut Imagine, AP-HP, Université de Paris, Paris, France
| | - Nicholas M Croft
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Johanna C Escher
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Lissy de Ridder
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.
| |
Collapse
|
5
|
Wang R, Wu D, Wang C, Livingston A, Wu X, Liu M, Yang XO. Platelet-Sourced TGF-β Promotes Th17 Responses and Enhances Airway Neutrophilia. Biomolecules 2025; 15:482. [PMID: 40305199 PMCID: PMC12024734 DOI: 10.3390/biom15040482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
Microbial, especially fungal, sensitization has been associated with the development and exacerbation of treatment-refractory neutrophilic asthma. Among the airway-inhabiting fungi, Aspergillus fumigatus and Candida albicans are the dominant species that elicit protective T helper (Th) 17 and other T cell responses, contributing to airway neutrophilia and steroid resistance. However, it is not fully understood how fungal airway colonization impacts the immunopathogenesis of asthma. Here, we used a neutrophilic asthma model induced by C. albicans to study the immune regulation of this disease. We found that intranasal administration of C. albicans induced platelet infiltration into the lung. Platelet-expressed latent TGF-β could be activated specifically by Th17 cells and drive the commitment, maintenance, and expansion of Th17 cells. In Candida-induced asthma, an adoptive transfer of platelets enhanced Th17 responses, increasing airway neutrophil influx. Thus, managing airway mycobiota and reducing platelet intrapulmonary infiltration may serve as a promising interventional approach.
Collapse
Affiliation(s)
- Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
| | - Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
| | - Chunqing Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (C.W.); (M.L.)
| | - Amanda Livingston
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (C.W.); (M.L.)
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (R.W.); (D.W.); (A.L.); (X.W.)
| |
Collapse
|
6
|
Parween F, Singh SP, Kathuria N, Zhang HH, Ashida S, Otaizo-Carrasquero FA, Shamsaddini A, Gardina PJ, Ganesan S, Kabat J, Lorenzi HA, Riley DJ, Myers TG, Pittaluga S, Bielekova B, Farber JM. Migration arrest and transendothelial trafficking of human pathogenic-like Th17 cells are mediated by differentially positioned chemokines. Nat Commun 2025; 16:1978. [PMID: 40000641 PMCID: PMC11861662 DOI: 10.1038/s41467-025-57002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Human Th17/type 17 cells express the chemokine receptor CCR6, but the functions of CCR6 and other chemokine receptors in human type 17 Th cell extravasation have not been fully delineated. Here we show that human peripheral blood CD4+CCR6+ T cells co-expressing CCR2 have a pathogenic Th17 signature, can produce inflammatory cytokines without T cell receptor activation, and show enhanced expression of pathogenicity-associated and activation-associated genes in the cerebrospinal fluid of patients with multiple sclerosis as compared to controls. In flow chambers with activated endothelial cell (EC) monolayers, CD4+CCR6+CCR2+ T cells are efficient at transendothelial migration (TEM). Ligands for CCR5, CCR6 and CXCR3 localize to EC surfaces and mediate only arrest, whereas CCR2 ligands fail to bind well to ECs and mediate only TEM. Conversely, expressing a chimeric CCR2 ligand engineered to bind glycosaminoglycans on ECs results in CCR2-mediated arrest but blocks TEM induction. Our results from human pathogenic-like type 17 cells thus suggest that T cell migration arrest requires chemokine bound to EC surfaces, whereas TEM requires a transendothelial chemokine gradient.
Collapse
Affiliation(s)
- Farhat Parween
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Satya P Singh
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nausheen Kathuria
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hongwei H Zhang
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shinji Ashida
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francisco A Otaizo-Carrasquero
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amirhossein Shamsaddini
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Paul J Gardina
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hernan A Lorenzi
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Deanna J Riley
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Timothy G Myers
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bibiana Bielekova
- Neuroimmunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joshua M Farber
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Kuklina EM. Mechanisms of Glucocorticoid Resistance in Nonclassical T Helper Populations Th17.1/Ex-Th17. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:188-199. [PMID: 40254398 DOI: 10.1134/s0006297924604222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 04/22/2025]
Abstract
The nonclassical population of Th1-polarized Th17 lymphocytes (Th17.1/ex-Th17) is currently in the focus of researchers' attention. These cells possess a unique proinflammatory potential and ability to penetrate blood-tissue barriers and play a key role in the pathogenesis of many inflammatory diseases, primarily autoimmune ones. Th1-polarized Th17 lymphocytes prevail in the autoimmune lesion foci and are considered to be a promising therapeutic target in these pathologies. At the same time, recent studies have shown another distinctive feature of Th1-polarized Th17 - their selective resistance to glucocorticoids. Since glucocorticoids are the first-line drugs for the treatment of the autoimmune disease exacerbation, understanding the causes of this phenomenon is crucial for predicting patients' response to therapy and improving the treatment effectiveness. This review analyzes the mechanisms of drug resistance of Th1-polarized Th17 cells, compares these mechanisms with those typical of nonpathogenic classical Th17 cells, and discusses the role of glucocorticoid resistance in the body's response to glucocorticoid therapy.
Collapse
Affiliation(s)
- Elena M Kuklina
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia.
| |
Collapse
|
8
|
Heredia M, Charrout M, Klomberg RCW, Aardoom MA, Jongsma MME, Kemos P, Hulleman-van Haaften DH, Tuk B, van Berkel LA, Bley Folly B, Calado B, Nugteren S, Simons-Oosterhuis Y, Doukas M, Sanders MA, van Beek G, Ruemmele FM, Croft NM, Mahfouz A, Reinders MJT, Escher JC, de Ridder L, Samsom JN. Combined plasma protein and memory T cell profiling discern IBD-patient-immunotypes related to intestinal disease and treatment outcomes. Mucosal Immunol 2025; 18:76-89. [PMID: 39332767 DOI: 10.1016/j.mucimm.2024.09.004] [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: 04/08/2024] [Revised: 08/29/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
Inflammatory bowel disease (IBD) chronicity results from memory T helper cell (Tmem) reactivation. Identifying patient-specific immunotypes is crucial for tailored treatment. We conducted a comprehensive study integrating circulating immune proteins and circulating Tmem, with intestinal tissue histology and mRNA analysis, in therapy-naïve pediatric IBD (Crohn's disease, CD: n = 62; ulcerative colitis, UC: n = 20; age-matched controls n = 43), and after 10-12 weeks' induction therapy. At diagnosis, plasma protein profiles unveiled two UC and three CD clusters with distinct disease courses. UC patients displayed unchanged circulating Tmem, while CD exhibited increased frequencies of gut-homing ex-Th17, known for high IFN-γ production. UC#2 had elevated Th17/neutrophil-pathway-related proteins and severe disease, with higher endoscopic and histological damage and Th17/neutrophil infiltration. Although both UC#1 and UC#2 responded to therapy, UC#2 required earlier immunomodulation. CD#3 had lower plasma protein concentrations, especially IFN-γ pathway proteins, fewer gut-homing ex-Th17 and clinically milder disease, confirmed by intestinal gene expression. CD#1 and CD#2 had comparably high Th1-related immune profiles, but CD#1 exhibited higher concentrations of proteins previously associated with poorer prognosis. Both CD clusters responded to induction therapy, with similar one-year outcomes. This study highlights feasibility of discriminating patient-specific immunotypes in IBD, advancing our understanding of immune pathogenesis, needed for tailored treatment strategies.
Collapse
Affiliation(s)
- Maud Heredia
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mohammed Charrout
- Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Renz C W Klomberg
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Martine A Aardoom
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Maria M E Jongsma
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Polychronis Kemos
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Danielle H Hulleman-van Haaften
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bastiaan Tuk
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lisette A van Berkel
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Brenda Bley Folly
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Beatriz Calado
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sandrine Nugteren
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ytje Simons-Oosterhuis
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mathijs A Sanders
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gregory van Beek
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Frank M Ruemmele
- Department of Pediatric Gastroenterology, Necker-Enfants Malades University Hospital, Institut Imagine, AP-HP, Université Paris Cité, Paris, France
| | - Nicholas M Croft
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Ahmed Mahfouz
- Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcel J T Reinders
- Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Johanna C Escher
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Lissy de Ridder
- Department of Pediatric Gastroenterology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
9
|
Harris EM, Chamseddine S, Chu A, Senkpeil L, Nikiciuk M, Bourdine A, Magin L, Al-Musa A, Woods B, Ozdogan E, Saker S, van Konijnenburg DPH, Yee CS, Nelson RW, Lee P, Halyabar O, Hale RC, Day-Lewis M, Henderson LA, Nguyen AA, Elkins M, Ohsumi TK, Gutierrez-Arcelus M, Peyper JM, Platt CD, Grace RF, LaBere B, Chou J. T cell and autoantibody profiling for primary immune regulatory disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.02.25.24303331. [PMID: 38464255 PMCID: PMC10925364 DOI: 10.1101/2024.02.25.24303331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Limited clinical tools exist for characterizing primary immune regulatory disorders (PIRD), which are often diagnoses of exclusion. Increased CD4+CXCR5+PD1+ circulating T follicular helper (cTfh) cell percentages have been identified as a marker of active disease in some, but not all, autoimmune disorders. Objective To develop a diagnostic approach that combines measurements of cellular and serologic autoimmunity. Methods We recruited 71 controls and 101 pediatric patients with PIRD with autoimmunity. Flow cytometry was used to measure CD4+CXCR5+ T cells expressing the chemokine receptors CXCR3 and/or CCR6. IgG and IgA autoantibodies were quantified in 56 patients and 20 controls using a microarray featuring 1616 full-length, conformationally intact protein antigens. The 97.5th percentile in the controls serves as the upper limit of normal for percentages of cTfh cells, CD4+CXCR5+ T cells expressing CXCR3 and/or CCR6, and autoantibody intensity and number. Results We found that 27.7% of patients had increased percentages of CD4+CXCR5+PD1+ cTfh cells and 42.5% had increased percentages of CD4+CXCR5+ cells expressing CXCR3 and/or CCR6. Patients had significantly more diverse IgG and IgA autoantibodies than controls and 37.5% had increased numbers of high-titer autoantibodies. Integrating measurements of cTfh cells, CD4+CXCR5+ T cells with CXCR3 and/or CCR6, and numbers of high-titer autoantibodies had 71.4% sensitivity (95% CI: 0.5852 - 0.8158) and 85% specificity (95% CI: 0.6396 - 0.9476) for patients with PIRD compared to controls. Conclusion By integrating CD4+ T cell phenotyping and total burden of autoantibodies, this approach provides additional tools for the diagnosis of PIRD lacking clinical diagnostic criteria.
Collapse
Affiliation(s)
- Emily M. Harris
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Sarah Chamseddine
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Anne Chu
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Northeastern University, Boston, MA
| | - Leetah Senkpeil
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew Nikiciuk
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Northeastern University, Boston, MA
| | - Aleksandra Bourdine
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Logan Magin
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Amer Al-Musa
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Woods
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Sarife Saker
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Christina S.K. Yee
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan W. Nelson
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Pui Lee
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Olha Halyabar
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca C. Hale
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan Day-Lewis
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lauren A. Henderson
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Alan A. Nguyen
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan Elkins
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Toshiro K. Ohsumi
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Craig D. Platt
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachael F. Grace
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Brenna LaBere
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Current affiliation: Division of Allergy and Immunology, Phoenix Children’s Hospital, Phoenix, AZ 85016
| | - Janet Chou
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Papanikolaou IC, Chytopoulos K, Kaitatzis D, Kostakis N, Bogiatzis A, Steiropoulos P, Drakopanagiotakis F. Phenotypes and Endotypes in Sarcoidosis: Unraveling Prognosis and Disease Course. Biomedicines 2025; 13:287. [PMID: 40002701 PMCID: PMC11853411 DOI: 10.3390/biomedicines13020287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Sarcoidosis is a multi-system granulomatous disease of unknown etiology. In genetically susceptible individuals, the precipitating factors generate, via immunity mechanisms, a host granulomatous response. The granuloma, for unknown reasons thus far, may resolve or may persist and lead to organ damage and fibrosis. Infectious agents, occupational exposure, obesity, smoking and genetic factors are implicated in the pathogenesis of sarcoidosis. Macrophages are important in granuloma formation, and their M1/M2 phenotype is associated with the prognosis of the disease. CD4+ T helper cells play a central role in the pathogenesis of sarcoidosis. The major contributors appear to be Th1 and Th17.1 cells, whose microenvironmental behavior is dictated by the secretions of macrophages and dendritic cells. Higher levels of Th1 and Th17.1 cells are associated with chronic disease and resistance to corticosteroid treatment. In recent years, advances in the phenotyping of sarcoidosis with the help of HRCT, PET-CT and lung function tests have provided us with a better understanding of the disease. Genetic phenotyping performed by the GenPhenReSa consortium and the SAGA study has led to the recognition of new, distinct phenotypes. The reconstitution of dysregulated autophagy through persistent m-TORC-1 pathways may be a new treatment target in sarcoidosis.
Collapse
Affiliation(s)
| | - Konstantinos Chytopoulos
- Department of Pneumonology, Medical School, Democritus University of Thrace, University General Hospital Dragana, 68100 Alexandroupolis, Greece (P.S.)
| | - Dimitrios Kaitatzis
- Department of Pneumonology, Medical School, Democritus University of Thrace, University General Hospital Dragana, 68100 Alexandroupolis, Greece (P.S.)
| | - Nikolaos Kostakis
- Department of Pneumonology, Medical School, Democritus University of Thrace, University General Hospital Dragana, 68100 Alexandroupolis, Greece (P.S.)
| | - Anastasios Bogiatzis
- Department of Pneumonology, Medical School, Democritus University of Thrace, University General Hospital Dragana, 68100 Alexandroupolis, Greece (P.S.)
| | - Paschalis Steiropoulos
- Department of Pneumonology, Medical School, Democritus University of Thrace, University General Hospital Dragana, 68100 Alexandroupolis, Greece (P.S.)
| | - Fotios Drakopanagiotakis
- Department of Pneumonology, Medical School, Democritus University of Thrace, University General Hospital Dragana, 68100 Alexandroupolis, Greece (P.S.)
| |
Collapse
|
11
|
Veltkamp SHC, Voorneveld PW. The Cell-Specific Effects of JAK1 Inhibitors in Ulcerative Colitis. J Clin Med 2025; 14:608. [PMID: 39860613 PMCID: PMC11766026 DOI: 10.3390/jcm14020608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
JAK1 inhibitors have become an important addition to the therapeutic options for ulcerative colitis (UC), targeting key inflammatory pathways mediated by cytokines such as the IL-6 family, interferons, IL-2 family, IL-10 family, and G-CSF. However, not all patients respond equally, and chronic inflammation persists in a subset of individuals. The variability in treatment response may reflect the heterogeneity of UC. Immune cells, epithelial cells, and stromal cells may have distinct contributions to disease pathogenesis. While JAK inhibitors were originally designed to target immune cells, their impact on non-immune cell types, such as epithelial and stromal cells, remains poorly understood. Investigating the mechanisms through which JAK1 inhibitors affect these diverse cellular populations and identifying the factors underlying differential responses is crucial to optimizing outcomes. This review explores the roles of immune, epithelial, and stromal cells in response to JAK1 inhibition and discusses potential strategies to improve treatment precision, such as predicting responders and identifying complementary therapeutic targets.
Collapse
Affiliation(s)
| | - Philip W. Voorneveld
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| |
Collapse
|
12
|
Ju X, Fard NE, Bhalla A, Dvorkin-Gheva A, Xiao M, Radford K, Zhang K, Ditta R, Oliveria JP, Paré G, Mukherjee M, Nair P, Sehmi R. A population of c-kit + IL-17A + ILC2s in sputum from individuals with severe asthma supports ILC2 to ILC3 trans-differentiation. Sci Transl Med 2025; 17:eado6649. [PMID: 39813318 DOI: 10.1126/scitranslmed.ado6649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/29/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
In prednisone-dependent severe asthma, uncontrolled sputum eosinophilia is associated with increased numbers of group 2 innate lymphoid cells (ILC2s). These cells represent a relatively steroid-insensitive source of interleukin-5 (IL-5) and IL-13 and are considered critical drivers of asthma pathology. The abundance of ILC subgroups in severe asthma with neutrophilic or mixed granulocytic (both eosinophilic and neutrophilic) airway inflammation, prone to recurrent infective exacerbations, remains unclear. Here, we found by flow cytometry that sputum ILC3s are increased in severe asthma with intense airway neutrophilia, whereas equivalently raised sputum ILC2s and ILC3s were found in severe asthma with mixed granulocytic inflammation. Unbiased clustering analyses identified an "intermediate-ILC2" population displaying markers of both ILC2s (prostaglandin D2 receptor 2; CRTH2, IL-5, and IL-13) and ILC3s (c-kit and IL-17A) that were most abundant in severe asthma with mixed granulocytic airway inflammation. Intermediate ILC2s correlated with airway neutrophilia and were associated with increased amounts of IL-1β and IL-18 in sputum supernatants. Coculture of sort-purified canonical ILC2s with IL-1β and IL-18 in vitro up-regulated c-kit and IL-17A as well as gene expression profiles related to both type 2 and type 17 inflammatory pathways. Together, we have identified an intermediate-ILC2 phenotype in the airways of individuals with severe mixed granulocytic asthma, representing a candidate therapeutic target for controlling neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Xiaotian Ju
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Nahal Emami Fard
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Anurag Bhalla
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Anna Dvorkin-Gheva
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Maria Xiao
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Katherine Radford
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Kayla Zhang
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Reina Ditta
- Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Clinical Research Laboratory and Biobank and the Genetic and Molecular Epidemiology Laboratory (CRLB-GMEL), Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada
| | - John Paul Oliveria
- Department of Biomarker Development, Genentech Inc., South San Francisco, CA 94080, USA
| | - Guillaume Paré
- Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Clinical Research Laboratory and Biobank and the Genetic and Molecular Epidemiology Laboratory (CRLB-GMEL), Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada
| | - Manali Mukherjee
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Roma Sehmi
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| |
Collapse
|
13
|
Chen Y, Liang R, Li Y, Jiang L, Ma D, Luo Q, Song G. Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application. Signal Transduct Target Ther 2024; 9:340. [PMID: 39627201 PMCID: PMC11615378 DOI: 10.1038/s41392-024-02030-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/04/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024] Open
Abstract
The dynamic regulation of chromatin accessibility is one of the prominent characteristics of eukaryotic genome. The inaccessible regions are mainly located in heterochromatin, which is multilevel compressed and access restricted. The remaining accessible loci are generally located in the euchromatin, which have less nucleosome occupancy and higher regulatory activity. The opening of chromatin is the most important prerequisite for DNA transcription, replication, and damage repair, which is regulated by genetic, epigenetic, environmental, and other factors, playing a vital role in multiple biological progresses. Currently, based on the susceptibility difference of occupied or free DNA to enzymatic cleavage, solubility, methylation, and transposition, there are many methods to detect chromatin accessibility both in bulk and single-cell level. Through combining with high-throughput sequencing, the genome-wide chromatin accessibility landscape of many tissues and cells types also have been constructed. The chromatin accessibility feature is distinct in different tissues and biological states. Research on the regulation network of chromatin accessibility is crucial for uncovering the secret of various biological processes. In this review, we comprehensively introduced the major functions and mechanisms of chromatin accessibility variation in different physiological and pathological processes, meanwhile, the targeted therapies based on chromatin dynamics regulation are also summarized.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Yong Li
- Hepatobiliary Pancreatic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lingli Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China.
| |
Collapse
|
14
|
Miedema J, Cinetto F, Smed-Sörensen A, Spagnolo P. The immunopathogenesis of sarcoidosis. J Autoimmun 2024; 149:103247. [PMID: 38734536 DOI: 10.1016/j.jaut.2024.103247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Sarcoidosis is a granulomatous multiorgan disease, thought to result from exposure to yet unidentified antigens in genetically susceptible individuals. The exaggerated inflammatory response that leads to granuloma formation is highly complex and involves the innate and adaptive immune system. Consecutive immunological studies using advanced technology have increased our understanding of aberrantly activated immune cells, mediators and pathways that influence the formation, maintenance and resolution of granulomas. Over the years, it has become increasingly clear that disease immunopathogenesis can only be understood if the clinical heterogeneity of sarcoidosis is taken into consideration, along with the distribution of immune cells in peripheral blood and involved organs. Most studies offer an immunological snapshot during disease course, while the cellular composition of both the circulation and tissue microenvironment may change over time. Despite these challenges, novel insights on the role of the immune system are continuously published, thus bringing the field forward. This review highlights current knowledge on the innate and adaptive immune responses involved in sarcoidosis pathogenesis, as well as the pathways involved in non-resolving disease and fibrosis development. Additionally, we describe proposed immunological mechanisms responsible for drug-induced sarcoid like reactions. Although many aspects of disease immunopathogenesis remain to be unraveled, the identification of crucial immune reactions in sarcoidosis may help identify new treatment targets. We therefore also discuss potential therapies and future strategies based on the latest immunological findings.
Collapse
Affiliation(s)
- Jelle Miedema
- Department of Pulmonary Medicine, Center of Expertise for Interstitial Lung Disease, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Italy; Department of Medicine - DIMED, University of Padova, Padova, Italy.
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy.
| |
Collapse
|
15
|
Thakare DR, Singh K, Qamar T, Singh D, Balakrishnan S, Rathore U, Jain N, Ora M, Misra DP. Serum p-Glycoprotein and Monomeric C-Reactive Protein are Elevated in Takayasu Arteritis. J Inflamm Res 2024; 17:8695-8712. [PMID: 39553310 PMCID: PMC11566576 DOI: 10.2147/jir.s490958] [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: 09/09/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Existing biomarkers including C-reactive protein (CRP) do not adequately distinguish active and inactive TAK. We compared serum p-glycoprotein (p-gp)/Multidrug Resistance Protein 1 (MDR1), monomeric CRP (mCRP), CRP, and mCRP:CRP ratio in Takayasu arteritis (TAK) and healthy controls and their relationship with disease activity. Patients and Methods Serum p-gp mCRP (ELISA) and CRP (nephelometry) were compared between consecutive adults with TAK (>18 years) enrolled from a prospective cohort (n = 92) and healthy controls (n = 29), and between active vs inactive TAK (n = 46 each). In a subset of active immunosuppressive-naïve TAK (n = 29), correlation was assessed between serum p-gp and p-gp expression on circulating T helper lymphocyte populations: overall (CD4+), Th17 (CD4+IL-17+), Th17.1 (CD4+IL-17+IFN-γ+) lymphocytes [normalized to Tregs (CD4+CD25+FoxP3+)]. Changes in serum p-gp, mCRP, CRP, and mCRP:CRP were compared before and after immunosuppression (n = 29). Data was represented using median (Q1-Q3). Receiver operating characteristics (ROC) curves were generated for TAK vs controls, and active vs inactive TAK with serum p-gp, mCRP, CRP, and mCRP:CRP. Multivariable-adjusted linear regression was used to predict active disease with serum p-gp, mCRP, CRP, or mCRP:CRP. Results Serum p-gp (11.19 vs 8.05 ng/mL), mCRP (1.61 vs 1.25 µg/L), and CRP (5.40 vs 2.1 mg/L) were elevated in TAK vs controls (p <0.05 for all). CRP was higher and mCRP:CRP ratio was lower in active vs inactive TAK (p < 0.001). ROC curves identified moderate prediction for active disease with CRP and inactive disease with serum p-gp (area under ROC curve 0.705 and 0.392, respectively). Multivariable-adjusted linear regression confirmed association of CRP with active disease (p = 0.009) and serum p-gp with inactive disease (p = 0.041). In treatment-naïve TAK, serum p-gp negatively correlated with p-gp+Th17.1 lymphocytes (Spearman's rho=-0.39, p = 0.046). CRP and serum p-gp were significantly lowered following immunosuppressive therapy in treatment-naïve TAK (p < 0.05). Conclusion Serum p-gp and mCRP are elevated in TAK. Serum p-gp is associated with inactive disease.
Collapse
Affiliation(s)
- Darpan Radheshyam Thakare
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
- Department of Clinical Immunology and Rheumatology, King George Medical University (KGMU), Lucknow, Uttar Pradesh, India
| | - Kritika Singh
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Tooba Qamar
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Deeksha Singh
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Sandeep Balakrishnan
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Upendra Rathore
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Neeraj Jain
- Department of Radiodiagnosis, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Manish Ora
- Department of Nuclear Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Durga Prasanna Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| |
Collapse
|
16
|
Ouboter LF, Lindelauf C, Jiang Q, Schreurs M, Abdelaal TR, Luk SJ, Barnhoorn MC, Hueting WE, Han-Geurts IJ, Peeters KCMJ, Holman FA, Koning F, van der Meulen-de Jong AE, Pascutti MF. Activated HLA-DR+CD38+ Effector Th1/17 Cells Distinguish Crohn's Disease-associated Perianal Fistulas from Cryptoglandular Fistulas. Inflamm Bowel Dis 2024; 30:2146-2161. [PMID: 38776553 PMCID: PMC11812577 DOI: 10.1093/ibd/izae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Indexed: 05/25/2024]
Abstract
BACKGROUND Perianal fistulas are a debilitating complication of Crohn's disease (CD). Due to unknown reasons, CD-associated fistulas are in general more difficult to treat than cryptoglandular fistulas (non-CD-associated). Understanding the immune cell landscape is a first step towards the development of more effective therapies for CD-associated fistulas. In this work, we characterized the composition and spatial localization of disease-associated immune cells in both types of perianal fistulas by high-dimensional analyses. METHODS We applied single-cell mass cytometry (scMC), spectral flow cytometry (SFC), and imaging mass cytometry (IMC) to profile the immune compartment in CD-associated perianal fistulas and cryptoglandular fistulas. An exploratory cohort (CD fistula, n = 10; non-CD fistula, n = 5) was analyzed by scMC to unravel disease-associated immune cell types. SFC was performed on a second fistula cohort (CD, n = 10; non-CD, n = 11) to comprehensively phenotype disease-associated T helper (Th) cells. IMC was used on a third cohort (CD, n = 5) to investigate the spatial distribution/interaction of relevant immune cell subsets. RESULTS Our analyses revealed that activated HLA-DR+CD38+ effector CD4+ T cells with a Th1/17 phenotype were significantly enriched in CD-associated compared with cryptoglandular fistulas. These cells, displaying features of proliferation, regulation, and differentiation, were also present in blood, and colocalized with other CD4+ T cells, CCR6+ B cells, and macrophages in the fistula tracts. CONCLUSIONS Overall, proliferating activated HLA-DR+CD38+ effector Th1/17 cells distinguish CD-associated from cryptoglandular perianal fistulas and are a promising biomarker in blood to discriminate between these 2 fistula types. Targeting HLA-DR and CD38-expressing CD4+ T cells may offer a potential new therapeutic strategy for CD-related fistulas.
Collapse
Affiliation(s)
- Laura F Ouboter
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ciska Lindelauf
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Qinyue Jiang
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mette Schreurs
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tamim R Abdelaal
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
- Systems and Biomedical Engineering Department, Faculty of Engineering Cairo University, Giza, Egypt
| | - Sietse J Luk
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Willem E Hueting
- Department of Surgery, Alrijne hospital, Leiderdorp, the Netherlands
| | | | - Koen C M J Peeters
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Fabian A Holman
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | |
Collapse
|
17
|
Vergara A, De Felice M, Cesaro A, Gragnano F, Pariggiano I, Golia E, De Pasquale A, Blasi E, Fimiani F, Monda E, Limongelli G, Calabrò P. Immune-Checkpoint Inhibitor-Related Myocarditis: Where We Are and Where We Will Go. Angiology 2024; 75:909-920. [PMID: 37699402 DOI: 10.1177/00033197231201929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Immune checkpoint inhibitors (ICIs) are specific monoclonal antibodies directed against inhibitory targets of the immune system, mainly represented by programmed death-1 (PD1) ligand-1 (PD-L1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4), thus enabling an amplified T-cell-mediated immune response against cancer cells. These drugs have significantly improved prognosis in patients with advanced metastatic cancer (e.g., melanoma, non-small cell lung cancer, renal cell carcinoma). However, uncontrolled activation of anti-tumor T-cells could trigger an excessive immune response, possibly responsible for multi-organ damage, including, among others, lymphocytic myocarditis. The incidence of ICIs-induced myocarditis is underestimated and the patients affected are poorly characterized. The diagnosis and management of this condition are mainly based on expert opinion and case reports. EKG and ultrasound are tests that can help identify patients at risk of myocarditis during treatment by red flags, such as QRS complex enlargement and narrowing of global longitudinal strain (GLS). Therapy of ICI-related myocarditis is based on immunosuppressors, monoclonal antibodies and fusion proteins. A future strategy could involve the use of microRNAs. This review considers the current state of the art of immune-related adverse cardiovascular events, focusing on histological and clinical features, diagnosis and management, including current treatments and future pharmacological targets.
Collapse
Affiliation(s)
- Andrea Vergara
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Marco De Felice
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Caserta, Italy
- Division of Oncology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Ivana Pariggiano
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Enrica Golia
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Antonio De Pasquale
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Ettore Blasi
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Fabio Fimiani
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli "V. Monaldi", Naples, Italy
| | - Emanuele Monda
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli "V. Monaldi", Naples, Italy
| | - Giuseppe Limongelli
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli "V. Monaldi", Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| |
Collapse
|
18
|
Guo Y, Ashrafizadeh M, Tambuwala MM, Ren J, Orive G, Yu G. P-glycoprotein (P-gp)-driven cancer drug resistance: biological profile, non-coding RNAs, drugs and nanomodulators. Drug Discov Today 2024; 29:104161. [PMID: 39245345 DOI: 10.1016/j.drudis.2024.104161] [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: 06/02/2024] [Revised: 08/07/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Drug resistance has compromised the efficacy of chemotherapy. The dysregulation of drug transporters including P-glycoprotein (P-gp) can mediate drug resistance through drug efflux. In this review, we highlight the role of P-gp in cancer drug resistance and the related molecular pathways, including phosphoinositide 3-kinase (PI3K)-Akt, phosphatase and tensin homolog (PTEN) and nuclear factor-κB (NF-κB), along with non-coding RNAs (ncRNAs). Extracellular vesicles secreted by the cells can transport ncRNAs and other proteins to change P-gp activity in cancer drug resistance. P-gp requires ATP to function, and the induction of mitochondrial dysfunction or inhibition of glutamine metabolism can impair P-gp function, thus increasing chemosensitivity. Phytochemicals, small molecules and nanoparticles have been introduced as P-gp inhibitors to increase drug sensitivity in human cancers.
Collapse
Affiliation(s)
- Yang Guo
- Department of Respiratory and Critical Care Medicine, Shenyang Tenth People's Hospital (Shenyang Chest Hospital), No. 11 Beihai Street, Dadong District, Shenyang 110044, Liaoning, China
| | - Milad Ashrafizadeh
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore.
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, No. 163 Shoushan Road, Jiangyin, China.
| |
Collapse
|
19
|
Tan KBC, Alexander HD, Linden J, Murray EK, Gibson DS. Anti-inflammatory effects of phytocannabinoids and terpenes on inflamed Tregs and Th17 cells in vitro. Exp Mol Pathol 2024; 139:104924. [PMID: 39208564 DOI: 10.1016/j.yexmp.2024.104924] [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: 01/09/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
AIMS Phytocannabinoids and terpenes from Cannabis sativa have demonstrated limited anti-inflammatory and analgesic effects in several inflammatory conditions. In the current study, we test the hypothesis that phytocannabinoids exert immunomodulatory effects in vitro by decreasing inflammatory cytokine expression and activation. KEY METHODS CD3/CD28 and lipopolysaccharide activated peripheral blood mononuclear cells (PBMCs) from healthy donors (n = 6) were treated with phytocannabinoid compounds and terpenes in vitro. Flow cytometry was used to determine regulatory T cell (Treg) and T helper 17 (Th17) cell responses to treatments. Cell pellets were harvested for qRT-PCR gene expression analysis of cytokines, cell activation markers, and inflammation-related receptors. Cell culture supernatants were analysed by ELISA to quantify IL-6, TNF-α and IL-10 secretion. MAIN FINDINGS In an initial screen of 20 μM cannabinoids and terpenes which were coded to blind investigators, cannabigerol (GL4a), caryophyllene oxide (GL5a) and gamma-terpinene (GL6a) significantly reduced cytotoxicity and gene expression levels of IL6, IL10, TNF, TRPV1, CNR1, HTR1A, FOXP3, RORC and NFKΒ1. Tetrahydrocannabinol (GL7a) suppression of T cell activation was associated with downregulation of RORC and NFKΒ1 gene expression and reduced IL-6 (p < 0.0001) and IL10 (p < 0.01) secretion. Cannabidiol (GL1b) significantly suppressed activation of Tregs (p < 0.05) and Th17 cells (p < 0.05) in a follow-on in vitro dose-response study. IL-6 (p < 0.01) and IL-10 (p < 0.01) secretion was significantly reduced with 50 μM cannabidiol. SIGNIFICANCE The study provides the first evidence that cannabidiol and tetrahydrocannabinol suppress extracellular expression of both anti- and pro-inflammatory cytokines in an in vitro PBMC model of inflammation.
Collapse
Affiliation(s)
- Kyle B C Tan
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Londonderry BT47 6SB, United Kingdom
| | - H Denis Alexander
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Londonderry BT47 6SB, United Kingdom
| | - James Linden
- GreenLight Pharmaceuticals Ltd, Unit 2, Block E, Nutgrove Office Park, Dublin 14, Ireland
| | - Elaine K Murray
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Londonderry BT47 6SB, United Kingdom
| | - David S Gibson
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Londonderry BT47 6SB, United Kingdom.
| |
Collapse
|
20
|
Sun N, Zhang M, Kong J, Li J, Dong Y, Wang X, Fu L, Zhou Y, Chen Y, Li Y, Sun X, Guo R. Dysregulated T-cell homeostasis and decreased CD30 + Treg proliferating in aplastic anemia. Heliyon 2024; 10:e35775. [PMID: 39170389 PMCID: PMC11337026 DOI: 10.1016/j.heliyon.2024.e35775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Aplastic anemia (AA) is an autoimmune hematopoietic disease mediated by autoreactive T cells leading to bone marrow failure. However, the precise role of autoreactive T cells in the development of AA is not fully understood, hindering the advancement of therapeutic and diagnostic strategies. In this study, we conducted a single-cell transcriptome analysis of CD8+ T cells, conventional CD4+ T (CD4+ Tconv) cells, and Treg cells, to elucidate the potential disruption of T cell homeostasis in patients with AA. We identified changes in CD4+ Tconv cells, including loss of homeostasis in naïve and memory cells and increased differentiation potential in T helper type 1 (TH1), T helper type 2 (TH2), and T helper type 17 (TH17) cells. Additionally, we identified naïve and memory CD8+ T cells that were enforced into an effector state. CD127 is an ideal surface marker for assessing the immune state of CD8+ T cells,as identified by flow cytometry. Abnormal expression of TNFSF8 has been observed in AA and other autoimmune diseases. Flow cytometry analysis revealed that TNFRSF8 (CD30), a receptor for TNFSF8, was predominantly present in human Treg cells. Importantly, patients with AA have a decreased CD30+ Treg subset. RNA-sequencing analysis revealed, that the CD30+ Treg cells are characterized by high proliferation and a remarkable immunosuppressive phenotype. Taken, together, we propose that abnormal TNFSF8/TNFRSF8 signaling is involved in dysfunctional T cell immunity by increasing the destruction of CD30+ Treg cells.
Collapse
Affiliation(s)
- Nannan Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengmeng Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Kong
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jin Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yong Dong
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaoqian Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liyan Fu
- Department of Laboratory Medicine, The First Clinical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yiwei Zhou
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaoyao Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianlei Sun
- Basic Medical Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
21
|
Punithavathy PM, Telugu RB, Rao VM, Prabhu SB, Kabeerdoss J, Syed C, Joseph G, Danda D, Thomas M, Goel R. Study of pathogenic T-helper cell subsets in Asian Indian patients with Takayasu arteritis. Immunol Res 2024; 72:636-643. [PMID: 38326692 DOI: 10.1007/s12026-024-09459-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
The relapses and refractory disease are a challenge in the management of patients with Takayasu arteritis (TAK). We quantified pathogenic CD4 + memory T helper cells bearing surface markers CD161 and/or p-glycoprotein (MDR1) in patients with TAK. Peripheral blood mononuclear cells of 21 patients with TAK and 16 age-matched controls were stained with anti-CD3, anti-CD4, anti-CD45RA, anti-CD161 and anti-p-glycoprotein antibodies and subjected to flow cytometry by FACS ARIAIII. Eighteen patients underwent follow-up immunophenotyping. Intracellular staining for interleukin-17 and interferon-γ was performed for 18 patients and 11 controls. Surgical arterial biopsies of 6 TAK and 5 non-inflammatory controls were subjected to immunohistochemistry with anti-CD161 and anti-p-glycoprotein. At baseline the frequency of MDR1 + CD4 + and CD161 + MDR1 + CD4 + memory T cells was higher in TAK than controls (p = 0.002 and 0.01, respectively). After stimulation, the frequency of IFN-y + CD161 + cells was higher in TAK than controls (p = 0.028). Modal fluorescence intensity of CD161 + MDR1 + CD45RA - CD4 + cells was higher in active as compared with stable disease (p = 0.041). At 6 months, MDR1 + and CD161 + MDR1 + memory CD4 + T cells decreased significantly only in patients who had complete/partial response to treatment (p = 0.047 and 0.02, respectively). To conclude, MDR1 + and MDR1 + CD161 + CD4 + memory T-helper cells are increased in patients with TAK. These cells decreased only in patients with response to treatment during subsequent follow-up.
Collapse
Affiliation(s)
- P M Punithavathy
- Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, 632004, India
| | - Ramesh Babu Telugu
- Department of Pathology, Christian Medical College, Vellore, 632004, India
| | - Vinay Murahari Rao
- Department of Cardiothoracic Surgery, Christian Medical College, Vellore, 632004, India
| | - Savit B Prabhu
- Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, 632004, India
| | - Jayakanthan Kabeerdoss
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, 632004, India
| | - Chanduni Syed
- Wellcome Trust Research Laboratory, Division of Gastrointestinal Sciences, Christian Medical College, Vellore, 632004, India
| | - George Joseph
- Department of Cardiology, Christian Medical College, Vellore, 632004, India
| | - Debashish Danda
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, 632004, India
| | - Meera Thomas
- Department of Pathology, Christian Medical College, Vellore, 632004, India
| | - Ruchika Goel
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, 632004, India.
| |
Collapse
|
22
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Campen M, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances Th17 Responses and Promotes Airway Neutrophilia. Am J Respir Cell Mol Biol 2024; 71:169-181. [PMID: 38593442 PMCID: PMC11299091 DOI: 10.1165/rcmb.2023-0424oc] [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: 12/01/2023] [Accepted: 04/09/2024] [Indexed: 04/11/2024] Open
Abstract
Heightened unfolded protein responses (UPRs) are associated with the risk for asthma, including severe asthma. Treatment-refractory severe asthma manifests a neutrophilic phenotype with T helper (Th)17 responses. However, how UPRs participate in the deregulation of Th17 cells leading to neutrophilic asthma remains elusive. This study found that the UPR sensor IRE1 is induced in the murine lung with fungal asthma and is highly expressed in Th17 cells relative to naive CD4+ T cells. Cytokine (e.g., IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by both human and mouse Th17 cells. Ern1 (encoding IRE1) deficiency decreases the expression of endoplasmic reticulum stress factors and impairs the differentiation and cytokine secretion of Th17 cells. Genetic ablation of Ern1 leads to alleviated Th17 responses and airway neutrophilia in a fungal airway inflammation model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances Th17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPR-mediated secretory function of Th17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in Th17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology and
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, and
| | | | - Ruoning Wang
- Department of Molecular Genetics and Microbiology and
| | | | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas; and
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology and
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Matthew Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | | | - Meilian Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, and
| | | |
Collapse
|
23
|
Guo S, Tian Y, Li J, Zeng X. A Glimpse into Humoral Response and Related Therapeutic Approaches of Takayasu's Arteritis. Int J Mol Sci 2024; 25:6528. [PMID: 38928233 PMCID: PMC11203527 DOI: 10.3390/ijms25126528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Takayasu's arteritis (TAK) manifests as an insidiously progressive and debilitating form of granulomatous inflammation including the aorta and its major branches. The precise etiology of TAK remains elusive, with current understanding suggesting an autoimmune origin primarily driven by T cells. Notably, a growing body of evidence bears testimony to the widespread effects of B cells on disease pathogenesis and progression. Distinct alterations in peripheral B cell subsets have been described in individuals with TAK. Advancements in technology have facilitated the identification of novel autoantibodies in TAK. Moreover, emerging data suggest that dysregulated signaling cascades downstream of B cell receptor families, including interactions with innate pattern recognition receptors such as toll-like receptors, as well as co-stimulatory molecules like CD40, CD80 and CD86, may result in the selection and proliferation of autoreactive B cell clones in TAK. Additionally, ectopic lymphoid neogenesis within the aortic wall of TAK patients exhibits functional characteristics. In recent decades, therapeutic interventions targeting B cells, notably utilizing the anti-CD20 monoclonal antibody rituximab, have demonstrated efficacy in TAK. Despite the importance of the humoral immune response, a systematic understanding of how autoreactive B cells contribute to the pathogenic process is still lacking. This review provides a comprehensive overview of the biological significance of B cell-mediated autoimmunity in TAK pathogenesis, as well as insights into therapeutic strategies targeting the humoral response. Furthermore, it examines the roles of T-helper and T follicular helper cells in humoral immunity and their potential contributions to disease mechanisms. We believe that further identification of the pathogenic role of autoimmune B cells and the underlying regulation system will lead to deeper personalized management of TAK patients. We believe that further elucidation of the pathogenic role of autoimmune B cells and the underlying regulatory mechanisms holds promise for the development of personalized approaches to managing TAK patients.
Collapse
Affiliation(s)
- Shuning Guo
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Yixiao Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| |
Collapse
|
24
|
Miller-Little WA, Chen X, Salazar V, Liu C, Bulek K, Zhou JY, Li X, Stüve O, Stappenbeck T, Dubyak G, Zhao J, Li X. A T H17-intrinsic IL-1β-STAT5 axis drives steroid resistance in autoimmune neuroinflammation. Sci Immunol 2024; 9:eabq1558. [PMID: 38701190 DOI: 10.1126/sciimmunol.abq1558] [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: 03/24/2022] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
Steroid resistance poses a major challenge for the management of autoimmune neuroinflammation. T helper 17 (TH17) cells are widely implicated in the pathology of steroid resistance; however, the underlying mechanisms are unknown. In this study, we identified that interleukin-1 receptor (IL-1R) blockade rendered experimental autoimmune encephalomyelitis (EAE) mice sensitive to dexamethasone (Dex) treatment. Interleukin-1β (IL-1β) induced a signal transducer and activator of transcription 5 (STAT5)-mediated steroid-resistant transcriptional program in TH17 cells, which promoted inflammatory cytokine production and suppressed Dex-induced anti-inflammatory genes. TH17-specific deletion of STAT5 ablated the IL-1β-induced steroid-resistant transcriptional program and rendered EAE mice sensitive to Dex treatment. IL-1β synergized with Dex to promote the STAT5-dependent expression of CD69 and the development of central nervous system (CNS)-resident CD69+ TH17 cells. Combined IL-1R blockade and Dex treatment ablated CNS-resident TH17 cells, reduced EAE severity, and prevented relapse. CD69+ tissue-resident TH17 cells were also detected in brain lesions of patients with multiple sclerosis. These findings (i) demonstrate that IL-1β-STAT5 signaling in TH17 cells mediates steroid resistance and (ii) identify a therapeutic strategy for reversing steroid resistance in TH17-mediated CNS autoimmunity.
Collapse
Affiliation(s)
- William A Miller-Little
- Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Xing Chen
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Vanessa Salazar
- Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Caini Liu
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Katarzyna Bulek
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Julie Y Zhou
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Xiao Li
- Center for RNA Science and Therapeutics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Olaf Stüve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Neurology Section, VA North Texas Health Care System, Medical Service Dallas, Veterans Affairs Medical Center, Dallas, TX, USA
| | - Thaddeus Stappenbeck
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - George Dubyak
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Junjie Zhao
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| |
Collapse
|
25
|
Wang X, He X, Liu J, Zhang H, Wan H, Luo J, Yang J. Immune pathogenesis of idiopathic granulomatous mastitis: from etiology toward therapeutic approaches. Front Immunol 2024; 15:1295759. [PMID: 38529282 PMCID: PMC10961981 DOI: 10.3389/fimmu.2024.1295759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
Idiopathic granulomatous mastitis (IGM) is a noncancerous, chronic inflammatory disorder of breast with unknown causes, posing significant challenges to the quality of life due to its high refractoriness and local aggressiveness. The typical symptoms of this disease involve skin redness, a firm and tender breast mass and mastalgia; others may include swelling, fistula, abscess (often without fever), nipple retraction, and peau d'orange appearance. IGM often mimics breast abscesses or malignancies, particularly inflammatory breast cancer, and is characterized by absent standardized treatment options, inconsistent patient response and unknown mechanism. Definite diagnosis of this disease relies on core needle biopsy and histopathological examination. The prevailing etiological theory suggests that IGM is an autoimmune disease, as some patients respond well to steroid treatment. Additionally, the presence of concurrent erythema nodosum or other autoimmune conditions supports the autoimmune nature of the disease. Based on current knowledge, this review aims to elucidate the autoimmune-favored features of IGM and explore its potential etiologies. Furthermore, we discuss the immune-mediated pathogenesis of IGM using existing research and propose immunotherapeutic strategies for managing this condition.
Collapse
Affiliation(s)
- Xiaoli Wang
- Breast Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiujing He
- Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Tumor Targeted and Immune Therapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Junzhi Liu
- West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Haiyan Zhang
- Department of Breast Surgery, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Hangyu Wan
- Department of Breast Surgery, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Jing Luo
- Department of Breast Surgery, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Jiqiao Yang
- Breast Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Padilla CM, Valenzi E, Tabib T, Nazari B, Sembrat J, Rojas M, Fuschiotti P, Lafyatis R. Increased CD8+ tissue resident memory T cells, regulatory T cells and activated natural killer cells in systemic sclerosis lungs. Rheumatology (Oxford) 2024; 63:837-845. [PMID: 37310903 PMCID: PMC10907815 DOI: 10.1093/rheumatology/kead273] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 06/15/2023] Open
Abstract
OBJECTIVE Multiple observations indicate a role for lymphocytes in driving autoimmunity in SSc. While T and NK cells have been studied in SSc whole blood and bronchoalveolar lavage fluid, their role remains unclear, partly because no studies have analysed these cell types in SSc-interstitial lung disease (ILD) lung tissue. This research aimed to identify and analyse the lymphoid subpopulations in SSc-ILD lung explants. METHODS Lymphoid populations from 13 SSc-ILD and 6 healthy control (HC) lung explants were analysed using Seurat following single-cell RNA sequencing. Lymphoid clusters were identified by their differential gene expression. Absolute cell numbers and cell proportions in each cluster were compared between cohorts. Additional analyses were performed using pathway analysis, pseudotime and cell ligand-receptor interactions. RESULTS Activated CD16+ NK cells, CD8+ tissue resident memory T cells and Treg cells were proportionately higher in SSc-ILD compared with HC lungs. Activated CD16+ NK cells in SSc-ILD showed upregulated granzyme B, IFN-γ and CD226. Amphiregulin, highly upregulated by NK cells, was predicted to interact with epidermal growth factor receptor on several bronchial epithelial cell populations. Shifts in CD8+ T cell populations indicated a transition from resting to effector to tissue resident phenotypes in SSc-ILD. CONCLUSIONS SSc-ILD lungs show activated lymphoid populations. Activated cytotoxic NK cells suggest they may kill alveolar epithelial cells, while their expression of amphiregulin suggests they may also induce bronchial epithelial cell hyperplasia. CD8+ T cells in SSc-ILD appear to transition from resting to the tissue resident memory phenotype.
Collapse
Affiliation(s)
- Cristina M Padilla
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eleanor Valenzi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tracy Tabib
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Banafsheh Nazari
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John Sembrat
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, OH, USA
| | - Patrizia Fuschiotti
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
27
|
Hughes K, Conaway E, Blackwell E, Rout E, Yoshimoto J, Burnett R, Avery A. Canine T zone lymphoma is a tumor of mature, previously activated αβ T cells. Vet Immunol Immunopathol 2024; 269:110725. [PMID: 38359755 DOI: 10.1016/j.vetimm.2024.110725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/20/2024] [Accepted: 01/24/2024] [Indexed: 02/17/2024]
Abstract
T cell lymphomas are a diverse group of tumors found in both dogs and humans, originating from various normal T cell types. Identifying the origin of neoplastic lymphocytes can offer valuable insights into the pathogenesis and clinical behavior of these tumors. T zone lymphoma (TZL) in dogs is characterized by the absence of CD45 expression, a strong breed predilection, and its association with adult-onset demodicosis-a condition believed to be linked to immunosuppression. In this study, our aim was to employ transcriptomic and functional data to determine the normal counterpart of TZL. Identifying the normal counterpart may help us understand both how these tumors arise and explain their clinical behavior. Gene expression profiling using NanoString and RNA seq was used to compare the transcriptome between neoplastic T zone cells, normal canine T cells and publicly available gene sets using Gene Set Enrichment Analysis. Mitogen, anti-CD3 stimulation and PMA/ionomycin stimulation were used to assess T cell proliferation in vitro, and intracellular cytokine production was measured by flow cytometry. Gene expression profiling revealed that TZL is most likely derived from an activated or memory alpha-beta T cell but the cells do not fall cleanly into an effector subtype. TZL cells express CD4-specific transcription factors GATA3 and THPOK, even though TZL cells more commonly express CD8, or neither CD4 nor CD8. TZL cells produce high levels of interferon gamma and tumor necrosis factor alpha when stimulated, further supporting the hypothesis that they are derived from an antigen experienced T cell. TZL cells do not proliferate when stimulated through the T cell receptor but will divide when the T cell receptor is bypassed with PMA and ionomycin. The observation that these cells are derived from a mature, previously activated T cell is the first step in understanding the genesis of this unique T cell tumor.
Collapse
Affiliation(s)
- Kelly Hughes
- Clinical Hematopathology Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO 80523, United States
| | - Evan Conaway
- Clinical Hematopathology Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO 80523, United States
| | - Emily Blackwell
- Clinical Hematopathology Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO 80523, United States
| | - Emily Rout
- Clinical Hematopathology Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO 80523, United States
| | - Janna Yoshimoto
- Clinical Hematopathology Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO 80523, United States
| | - Robert Burnett
- Clinical Hematopathology Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO 80523, United States
| | - Anne Avery
- Clinical Hematopathology Laboratory, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, CO 80523, United States.
| |
Collapse
|
28
|
Petakh P, Oksenych V, Kamyshna I, Boisak I, Lyubomirskaya K, Kamyshnyi O. Exploring the complex interplay: gut microbiome, stress, and leptospirosis. Front Microbiol 2024; 15:1345684. [PMID: 38476949 PMCID: PMC10927737 DOI: 10.3389/fmicb.2024.1345684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/30/2024] [Indexed: 03/14/2024] Open
Abstract
Leptospirosis, a re-emerging zoonotic disease, remains a significant global health concern, especially amid floods and disasters such as the Kakhovka Dam destruction. As is known, the stress that occurs in the conditions of military conflicts among civilian and military personnel significantly affects susceptibility to infectious diseases and possibly even influences their course. This review aims to explore how the gut microbiome and stress mediators (such as catecholamines and corticosteroids) might impact the leptospirosis disease course. The review opens new horizons for research by elucidating the connections between the gut microbiome, stress, and leptospirosis.
Collapse
Affiliation(s)
- Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Boisak
- Department of Childhood Diseases, Uzhhorod National University, Uzhhorod, Ukraine
| | - Katerina Lyubomirskaya
- Department of Obstetrics and Gynecology, Zaporizhzhia State Medical and Pharmaceuticals University, Zaporizhzhia, Ukraine
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
29
|
Wen Y, Wang H, Tian D, Wang G. TH17 cell: a double-edged sword in the development of inflammatory bowel disease. Therap Adv Gastroenterol 2024; 17:17562848241230896. [PMID: 38390028 PMCID: PMC10883129 DOI: 10.1177/17562848241230896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 01/17/2024] [Indexed: 02/24/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic nonspecific inflammatory disease of the gastrointestinal tract, and its pathogenesis has not been fully understood. Extensive dysregulation of the intestinal mucosal immune system is critical in the development and progression of IBD. T helper (Th) 17 cells have the characteristics of plasticity. They can transdifferentiate into subpopulations with different functions in response to different factors in the surrounding environment, thus taking on different roles in regulating the intestinal immune responses. In this review, we will focus on the plasticity of Th17 cells as well as the function of Th17 cells and their related cytokines in IBD. We will summarize their pathogenic and protective roles in IBD under different conditions, respectively, hoping to further deepen the understanding of the pathological mechanisms underlying IBD and provide insights for future treatment.
Collapse
Affiliation(s)
- Yue Wen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ge Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
30
|
Hoeks C, Puijfelik FV, Koetzier SC, Rip J, Corsten CEA, Wierenga-Wolf AF, Melief MJ, Stinissen P, Smolders J, Hellings N, Broux B, van Luijn MM. Differential Runx3, Eomes, and T-bet expression subdivides MS-associated CD4 + T cells with brain-homing capacity. Eur J Immunol 2024; 54:e2350544. [PMID: 38009648 DOI: 10.1002/eji.202350544] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
Multiple sclerosis (MS) is a common and devastating chronic inflammatory disease of the CNS. CD4+ T cells are assumed to be the first to cross the blood-central nervous system (CNS) barrier and trigger local inflammation. Here, we explored how pathogenicity-associated effector programs define CD4+ T cell subsets with brain-homing ability in MS. Runx3- and Eomes-, but not T-bet-expressing CD4+ memory cells were diminished in the blood of MS patients. This decline reversed following natalizumab treatment and was supported by a Runx3+ Eomes+ T-bet- enrichment in cerebrospinal fluid samples of treatment-naïve MS patients. This transcription factor profile was associated with high granzyme K (GZMK) and CCR5 levels and was most prominent in Th17.1 cells (CCR6+ CXCR3+ CCR4-/dim ). Previously published CD28- CD4 T cells were characterized by a Runx3+ Eomes- T-bet+ phenotype that coincided with intermediate CCR5 and a higher granzyme B (GZMB) and perforin expression, indicating the presence of two separate subsets. Under steady-state conditions, granzyme Khigh Th17.1 cells spontaneously passed the blood-brain barrier in vitro. This was only found for other subsets including CD28- cells when using inflamed barriers. Altogether, CD4+ T cells contain small fractions with separate pathogenic features, of which Th17.1 seems to breach the blood-brain barrier as a possible early event in MS.
Collapse
Affiliation(s)
- Cindy Hoeks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Fabiënne van Puijfelik
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Steven C Koetzier
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jasper Rip
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Cato E A Corsten
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Annet F Wierenga-Wolf
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marie-José Melief
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Piet Stinissen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Niels Hellings
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Bieke Broux
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Center (UMSC), Hasselt, Belgium
| | - Marvin M van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
31
|
Martinez GJ, Appleton M, Kipp ZA, Loria AS, Min B, Hinds TD. Glucocorticoids, their uses, sexual dimorphisms, and diseases: new concepts, mechanisms, and discoveries. Physiol Rev 2024; 104:473-532. [PMID: 37732829 PMCID: PMC11281820 DOI: 10.1152/physrev.00021.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/07/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023] Open
Abstract
The normal stress response in humans is governed by the hypothalamic-pituitary-adrenal (HPA) axis through heightened mechanisms during stress, raising blood levels of the glucocorticoid hormone cortisol. Glucocorticoids are quintessential compounds that balance the proper functioning of numerous systems in the mammalian body. They are also generated synthetically and are the preeminent therapy for inflammatory diseases. They act by binding to the nuclear receptor transcription factor glucocorticoid receptor (GR), which has two main isoforms (GRα and GRβ). Our classical understanding of glucocorticoid signaling is from the GRα isoform, which binds the hormone, whereas GRβ has no known ligands. With glucocorticoids being involved in many physiological and cellular processes, even small disruptions in their release via the HPA axis, or changes in GR isoform expression, can have dire ramifications on health. Long-term chronic glucocorticoid therapy can lead to a glucocorticoid-resistant state, and we deliberate how this impacts disease treatment. Chronic glucocorticoid treatment can lead to noticeable side effects such as weight gain, adiposity, diabetes, and others that we discuss in detail. There are sexually dimorphic responses to glucocorticoids, and women tend to have a more hyperresponsive HPA axis than men. This review summarizes our understanding of glucocorticoids and critically analyzes the GR isoforms and their beneficial and deleterious mechanisms and the sexual differences that cause a dichotomy in responses. We also discuss the future of glucocorticoid therapy and propose a new concept of dual GR isoform agonist and postulate why activating both isoforms may prevent glucocorticoid resistance.
Collapse
Affiliation(s)
- Genesee J Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Malik Appleton
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
32
|
Kim JS, Gupta R. Clinical Manifestations and Management of Fibrotic Pulmonary Sarcoidosis. J Clin Med 2023; 13:241. [PMID: 38202248 PMCID: PMC10780222 DOI: 10.3390/jcm13010241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Fibrotic pulmonary sarcoidosis represents a distinct and relatively uncommon manifestation within the spectrum of sarcoidosis and has substantial morbidity and mortality. Due to the scarcity of research focused on this specific disease subtype, our current understanding of pathogenesis and optimal management remains constrained. This knowledge gap underscores the need for further investigation into areas such as targeted therapies, lung transplantation, and quality of life of patients with fibrotic pulmonary sarcoidosis. The primary aim of this review is to discuss recent developments within the realm of fibrotic pulmonary sarcoidosis to foster a more comprehensive understanding of the underlying mechanisms, prognosis, and potential treatment modalities.
Collapse
Affiliation(s)
- Jin Sun Kim
- Department of Thoracic Medicine and Surgery, Temple University Hospital, Philadelphia, PA 19140, USA
| | - Rohit Gupta
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University Hospital, Philadelphia, PA 19140, USA;
| |
Collapse
|
33
|
Paroni M, Leccese G, Ranzani V, Moschetti G, Chiara M, Perillo F, Ferri S, Clemente F, Noviello D, Conforti FS, Ferrero S, Karnani B, Bosotti R, Vasco C, Curti S, Crosti MC, Gruarin P, Rossetti G, Conte MP, Vecchi M, Pagani M, Landini P, Facciotti F, Abrignani S, Caprioli F, Geginat J. An Intestinal Th17 Subset is Associated with Inflammation in Crohn's Disease and Activated by Adherent-invasive Escherichia coli. J Crohns Colitis 2023; 17:1988-2001. [PMID: 37462681 PMCID: PMC10798865 DOI: 10.1093/ecco-jcc/jjad119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2024]
Abstract
IFNγ-producing ex-Th17 cells ['Th1/17'] were shown to play a key pathogenic role in experimental colitis and are abundant in the intestine. Here, we identified and characterised a novel, potentially colitogenic subset of Th17 cells in the intestine of patients with Crohn's disease [CD]. Human Th17 cells expressing CCR5 ['pTh17'] co-expressed T-bet and RORC/γt and produced very high levels of IL-17, together with IFN-γ. They had a gene signature of Th17 effector cells and were distinct from established Th1/17 cells. pTh17 cells, but not Th1/17 cells, were associated with intestinal inflammation in CD, and decreased upon successful anti-TNF therapy with infliximab. Conventional CCR5[-]Th17 cells differentiated to pTh17 cells with IL-23 in vitro. Moreover, anti-IL-23 therapy with risankizumab strongly reduced pTh17 cells in the intestine. Importantly, intestinal pTh17 cells were selectively activated by adherent-invasive Escherichia coli [AIEC], but not by a commensal/probiotic E. coli strain. AIEC induced high levels of IL-23 and RANTES from dendritic cells [DC]. Intestinal CCR5+Th1/17 cells responded instead to cytomegalovirus and were reduced in ulcerative colitis [UC], suggesting an unexpected protective role. In conclusion, we identified an IL-23-inducible subset of human intestinal Th17 cells. pTh17 cells produced high levels of pro-inflammatory cytokines, were selectively associated with intestinal inflammation in CD, and responded to CD-associated AIEC, suggesting a key colitogenic role.
Collapse
Affiliation(s)
- Moira Paroni
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Gabriella Leccese
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Valeria Ranzani
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Giorgia Moschetti
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Matteo Chiara
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Federica Perillo
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Sara Ferri
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Francesca Clemente
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Daniele Noviello
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Francesco Simone Conforti
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Pathology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Biomedical, Surgical, and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| | - Bhavna Karnani
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Roberto Bosotti
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Chiara Vasco
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Serena Curti
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Maria Cristina Crosti
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Paola Gruarin
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
| | - Grazisa Rossetti
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- Molecular Oncology and Immunology, FIRC Institute of Molecular Oncology [IFOM], Milan, Italy
| | - Maria Pia Conte
- Department of Public Health and Infectious Diseases, ‘Sapienza’ University of Rome, Rome, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Massimiliano Pagani
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- Molecular Oncology and Immunology, FIRC Institute of Molecular Oncology [IFOM], Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paolo Landini
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Federica Facciotti
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Sergio Abrignani
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- DISCCO, Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Jens Geginat
- INGM-National Institute of Molecular Genetics ‘Romeo ed Enrica Invernizzi’, Milan, Italy
- DISCCO, Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
34
|
Zielinski CE. T helper cell subsets: diversification of the field. Eur J Immunol 2023; 53:e2250218. [PMID: 36792132 DOI: 10.1002/eji.202250218] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Polarized T helper cell (Th cell) responses are important determinants of host protection. Th cell subsets tailor their functional repertoire of cytokines to their cognate antigens to efficiently contribute to their clearance. In contrast, in settings of immune abrogation, these polarized cytokine patterns of Th cells can mediate tissue damage and pathology resulting in allergy or autoimmunity. Recent technological developments in single-cell genomics and proteomics as well as advances in the high-dimensional bioinformatic analysis of complex datasets have challenged the prevailing Th cell subset classification into Th1, Th2, Th17, and other subsets. Additionally, systems immunology approaches have revealed that instructive input from the peripheral tissue microenvironment can have differential effects on the overall phenotype and molecular wiring of Th cells depending on their spatial distribution. Th cells from the blood or secondary lymphoid organs are therefore expected to follow distinct rules of regulation. In this review, the functional heterogeneity of Th cell subsets will be reviewed in the context of new technological developments and T-cell compartmentalization in tissue niches. This work will especially focus on challenges to the traditional boundaries of Th cell subsets and will discuss the underlying regulatory checkpoints, which could reveal new therapeutic strategies for various immune-mediated diseases.
Collapse
Affiliation(s)
- Christina E Zielinski
- Department of Infection Immunology, Leibniz Institute for Natural Products Research and Infection Biology, Jena, Germany
- Institute of Microbiology, Faculty of Biosciences, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
35
|
Khantakova JN, Mutovina A, Ayriyants KA, Bondar NP. Th17 Cells, Glucocorticoid Resistance, and Depression. Cells 2023; 12:2749. [PMID: 38067176 PMCID: PMC10706111 DOI: 10.3390/cells12232749] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Depression is a severe mental disorder that disrupts mood and social behavior and is one of the most common neuropsychological symptoms of other somatic diseases. During the study of the disease, a number of theories were put forward (monoamine, inflammatory, vascular theories, etc.), but none of those theories fully explain the pathogenesis of the disease. Steroid resistance is a characteristic feature of depression and can affect not only brain cells but also immune cells. T-helper cells 17 type (Th17) are known for their resistance to the inhibitory effects of glucocorticoids. Unlike the inhibitory effect on other subpopulations of T-helper cells, glucocorticoids can enhance the differentiation of Th17 lymphocytes, their migration to the inflammation, and the production of IL-17A, IL-21, and IL-23 in GC-resistant disease. According to the latest data, in depression, especially the treatment-resistant type, the number of Th17 cells in the blood and the production of IL-17A is increased, which correlates with the severity of the disease. However, there is still a significant gap in knowledge regarding the exact mechanisms by which Th17 cells can influence neuroinflammation in depression. In this review, we discuss the mutual effect of glucocorticoid resistance and Th17 lymphocytes on the pathogenesis of depression.
Collapse
Affiliation(s)
- Julia N. Khantakova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva 10, Novosibirsk 630090, Russia; (K.A.A.); (N.P.B.)
| | - Anastasia Mutovina
- Department of Natural Sciences, Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia;
| | - Kseniya A. Ayriyants
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva 10, Novosibirsk 630090, Russia; (K.A.A.); (N.P.B.)
| | - Natalia P. Bondar
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Lavrentyeva 10, Novosibirsk 630090, Russia; (K.A.A.); (N.P.B.)
- Department of Natural Sciences, Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia;
| |
Collapse
|
36
|
Gosser C, Al Bawaliz A, Bahaj W, Chesney J, Ranjan S. Immune Checkpoint Inhibitor-Induced Myositis/Myasthenia Gravis Overlap. Cureus 2023; 15:e49007. [PMID: 38111441 PMCID: PMC10726830 DOI: 10.7759/cureus.49007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2023] [Indexed: 12/20/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have considerably changed the management of several malignancies. Although these agents transformed the scope of management in oncology and proved long-term efficacy, they have been associated with numerous autoimmune-related adverse events. We presented a case of a 61-year-old male with a history of non-small cell lung cancer (NSCLC) who presented with respiratory failure requiring mechanical ventilation. He was discharged with a working diagnosis of myasthenia gravis crisis secondary to the use of pembrolizumab. On further evaluation, he was found to possibly have pembrolizumab-induced myositis. He was treated with plasmapheresis, methylprednisolone, and rituximab and achieved significant improvement. Pembrolizumab, a monoclonal antibody, is an ICI that targets programmed death protein 1 (PD-1), thereby blocking the interaction between PD-1 and PDL-1, leading to an enhancement of T-cell mediated immune response against tumor cells. Pembrolizumab has been used to treat a variety of malignancies including melanoma, NSCLC, and other solid tumors. Though ICIs have revolutionized the field of oncology, they should be used with caution. ICIs can cause immune-related adverse events (irAEs), including myasthenia gravis and myositis. Diagnosing irAEs is challenging due to their nonspecific presentations and lack of antibody markers. Therefore, patients and clinicians should be aware of irAEs in order to initiate timely intervention.
Collapse
Affiliation(s)
- Caroline Gosser
- Internal Medicine, University of Louisville School of Medicine, Louisville, USA
| | - Anas Al Bawaliz
- Hematology and Medical Oncology, University of Louisville Hospital, Louisville, USA
| | - Waled Bahaj
- Hematology and Medical Oncology, University of Louisville Hospital, Louisville, USA
| | - Jason Chesney
- Hematology and Medical Oncology, University of Louisville Hospital, Louisville, USA
| | - Smita Ranjan
- Hematology and Medical Oncology, University of Louisville Hospital, Louisville, USA
| |
Collapse
|
37
|
Di Roio A, Hubert M, Besson L, Bossennec M, Rodriguez C, Grinberg-Bleyer Y, Lalle G, Moudombi L, Schneider R, Degletagne C, Treilleux I, Campbell DJ, Metzger S, Duhen T, Trédan O, Caux C, Ménétrier-Caux C. MDR1-EXPRESSING CD4 + T CELLS WITH TH1.17 FEATURES RESIST TO NEOADJUVANT CHEMOTHERAPY AND ARE ASSOCIATED WITH BREAST CANCER CLINICAL RESPONSE. J Immunother Cancer 2023; 11:e007733. [PMID: 37940345 PMCID: PMC10632904 DOI: 10.1136/jitc-2023-007733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Multidrug resistance-1 (MDR1) transporter limits the intracellular accumulation of chemotherapies (paclitaxel, anthracyclines) used in breast cancer (BC) treatment. In addition to tumor cells, MDR1 is expressed on immune cell subsets in which it confers chemoresistance. Among human T cells, MDR1 is expressed by most CD8+ T cells, and a subset of CD4+ T helper (Th) cells. Here we explored the expression, function and regulation of MDR1 on CD4+ T cells and investigated the role of this population in response to neoadjuvant chemotherapy (NAC) in BC. METHODS Phenotypic and functional characteristics of MDR1+ CD4 Th cells were assessed on blood from healthy donors and patients with BC by flow cytometry. These features were extended to CD4+ Th cells from untreated breast tumor by flow cytometry and RNA-sequencing (RNA-seq). We performed in vitro polarization assays to decipher MDR1 regulation on CD4 Th cells. We evaluated in vitro the impact of chemotherapy agents on MDR1+ CD4+ Th cells. We analyzed the impact of NAC treatment on MDR1+ CD4+ Th cells from blood and tumors and their association with treatment efficacy in two independent BC cohorts and in a public RNA-seq data set of BC tumor biopsies before and after NAC. Finally, we performed single cell (sc) RNAseq of blood CD4+ memory T cells from NAC-treated patients and combined them with an scRNAseq public data set. RESULTS MDR1+ CD4 Th cells were strongly enriched in Th1.17 polyfunctional cells but also in Th17 cells, both in blood and untreated breast tumor tissues. Mechanistically, Tumor growth factor (TGF)-β1 was required for MDR1 induction during in vitro Th17 or Th1.17 polarization. MDR1 expression conferred a selective advantage to Th1.17 and Th17 cells following paclitaxel treatment in vitro and in vivo in NAC-treated patients. scRNAseq demonstrated MDR1 association with tumor Th1.17 and Th with features of cytotoxic cells. Enrichment in MDR1+ CD4+ Th1.17 and Th17 cells, in blood and tumors positively correlated with pathological response. Absence of early modulation of Th1.17 and Th17 in NAC-resistant patients, argue for its use as a biomarker for chemotherapy regimen adjustment. CONCLUSION MDR1 favored the enrichment of Th1.17 and Th17 in blood and tumor after NAC that correlated to clinical response.
Collapse
Affiliation(s)
- Anthony Di Roio
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | - Margaux Hubert
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | - Laurie Besson
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | - Marion Bossennec
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | - Céline Rodriguez
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | | | - Guilhem Lalle
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | - Lyvia Moudombi
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | - Raphael Schneider
- Plateforme Gilles Thomas, Centre de Recherche en cancérologie de Lyon, Lyon, France
| | - Cyril Degletagne
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | - Isabelle Treilleux
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
- BioPathology Department, Centre Léon Bérard, Lyon, Rhône-Alpes, France
| | - Daniel J Campbell
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Séverine Metzger
- Clinical Research Platform, DRCI, Centre Léon Bérard, Lyon, Rhône-Alpes, France
| | - Thomas Duhen
- Earle A Chiles Research Institute, Portland, Oregon, USA
| | | | - Christophe Caux
- TERI Department, Centre de Recherche en Cancerologie de Lyon, Lyon, France
| | | |
Collapse
|
38
|
Balasubramanian A, Sundrud MS. ATP-dependent transporters: emerging players at the crossroads of immunity and metabolism. Front Immunol 2023; 14:1286696. [PMID: 38022644 PMCID: PMC10644303 DOI: 10.3389/fimmu.2023.1286696] [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: 08/31/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Nearly 50 ATP-binding cassette (ABC) transporters are encoded by mammalian genomes. These transporters are characterized by conserved nucleotide-binding and hydrolysis (i.e., ATPase) domains, and power directional transport of diverse substrate classes - ions, small molecule metabolites, xenobiotics, hydrophobic drugs, and even polypeptides - into or out of cells or subcellular organelles. Although immunological functions of ABC transporters are only beginning to be unraveled, emerging literature suggests these proteins have under-appreciated roles in the development and function of T lymphocytes, including many of the key effector, memory and regulatory subsets that arise during responses to infection, inflammation or cancers. One transporter in particular, MDR1 (Multidrug resistance-1; encoded by the ABCB1 locus in humans), has taken center stage as a novel player in immune regulation. Although MDR1 remains widely viewed as a simple drug efflux pump in tumor cells, recent evidence suggests that this transporter fills key endogenous roles in enforcing metabolic fitness of activated CD4 and CD8 T cells. Here, we summarize current understanding of the physiological functions of ABC transporters in immune regulation, with a focus on the anti-oxidant functions of MDR1 that may shape both the magnitude and repertoires of antigen-specific effector and memory T cell compartments. While much remains to be learned about the functions of ABC transporters in immunobiology, it is already clear that they represent fertile new ground, both for the definition of novel immunometabolic pathways, and for the discovery of new drug targets that could be leveraged to optimize immune responses to vaccines and cancer immunotherapies.
Collapse
Affiliation(s)
- Akshaya Balasubramanian
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Mark S. Sundrud
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Center for Digestive Health, Dartmouth Health, Lebanon, NH, United States
- Dartmouth Cancer Center, Lebanon, NH, United States
| |
Collapse
|
39
|
Jovanovic M, Sabovic M. Refractory drug-induced systemic small-vessel vasculitis with two varied extracutaneous manifestations: a case report and review of the literature. J Med Case Rep 2023; 17:470. [PMID: 37885023 PMCID: PMC10605860 DOI: 10.1186/s13256-023-04174-8] [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: 06/05/2023] [Accepted: 09/11/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Clopidogrel and ticagrelor are rarely reported to cause vasculitis via drug hypersensitivity reaction, largely mediated by T cells and immunoglobulin E (IgE). Despite therapeutic advances, the etiology of refractory vasculitides remains incompletely understood. Recently, (non)immunological mechanisms bypassing T cells and IgE have been proposed to explain resistance to standard immunosuppressants. Herein, we report a case of refractory drug-induced systemic small-vessel vasculitis with varied extracutaneous manifestations and incorporate multiple sources of data to provide detailed accounts of complex (non)immunological phenomena involved in this case. Study objectives are to provide an insight about rare presentations of commonly used drugs, upgrade the pathophysiological concepts of drug-induced vasculitis, raise need for further investigation to define causes and risk factors for refractory vasculitis, and discuss most of the current knowledge suggesting novel therapeutic approaches to treat this vasculitis. To our knowledge, this is the first case of the two flares of systemic small-vessel vasculitis in a single patient in response to clopidogrel and ticagrelor exposure, respectively. However, this report is limited by attribution/observer bias. CASE PRESENTATION We herein report a 24-year-old Caucasian male student with a medical history of mild seasonal allergic rhinoconjunctivitis, tension-type headaches, posttraumatic arterial stenosis, and previous exposure to ibuprofen, acetylsalicylic acid, and mRNA coronavirus disease 2019 (COVID-19) vaccine who suffered largely from acute urticaria and dyspnea after 20 days of acetylsalicylic acid and clopidogrel introduction. A skin punch biopsy confirmed leukocytoclastic vasculitis. Serologic antibody testing, complement analysis, microbiologic testing, and cancer biomarkers revealed no abnormalities. Regarding the patient's medical history, both acetylsalicylic acid and clopidogrel were exchanged for ticagrelor. Furthermore, the addition of naproxen, cyclosporine, bilastine, prednisolone, and montelukast resulted in complete recovery. After 7 days, diarrhea and hematuria occurred. Urinalysis and computed tomography showed reversible proteinuria with gross hematuria and hypodense changes in kidney medulla, respectively, associated with discontinuation of ticagrelor and naproxen. In addition, the patient recovered completely without any immunosuppression up-titration. CONCLUSIONS This case highlights the role of clopidogrel and ticagrelor as possible triggering agents for systemic small-vessel vasculitis and offers an insight into novel therapeutic strategies for refractory vasculitides. Further research is needed to build on the findings of a current report.
Collapse
Affiliation(s)
- Mark Jovanovic
- Department of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia.
| | - Miso Sabovic
- Department of Angiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
40
|
Toghi M, Bitarafan S, Ghafouri-Fard S. Pathogenic Th17 cells in autoimmunity with regard to rheumatoid arthritis. Pathol Res Pract 2023; 250:154818. [PMID: 37729783 DOI: 10.1016/j.prp.2023.154818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Th17 cells contribute the pathobiology of autoimmune diseases, including rheumatoid arthritis (RA). However, it was shown that differentiated Th17 cells display a high degree of plasticity under the influence of inflammatory conditions. In some autoimmune diseases, the majority of Th17 cells, especially at sites of inflammation, have a phenotype that is intermediate between Th17 and Th1. These cells, which are described as Th17.1 or exTh17 cells, are hypothesized to be more pathogenic than classical Th17 cells. In this review, the involvement of Th17.1 lymphocytes in RA, and potential features that might render these cells to be more pathogenic are discussed.
Collapse
Affiliation(s)
- Mehdi Toghi
- Department of Immune and Infectious Diseases, Université Laval, Quebec City, Quebec, Canada
| | - Sara Bitarafan
- Department of Molecular Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Vale N, Pereira M, Mendes RA. Systemic Inflammatory Disorders, Immunosuppressive Treatment and Increase Risk of Head and Neck Cancers-A Narrative Review of Potential Physiopathological and Biological Mechanisms. Cells 2023; 12:2192. [PMID: 37681925 PMCID: PMC10487135 DOI: 10.3390/cells12172192] [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: 07/22/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
Head and neck cancers (HNCs) are known to present multiple factors likely to influence their development. This review aims to provide a comprehensive overview of the current scientific literature on the interplay between systemic inflammatory disorders, immunosuppressive treatments and their synergistic effect on HNC risk. Both cell-mediated and humoral-mediated systemic inflammatory disorders involve dysregulated immune responses and chronic inflammation and these inflammatory conditions have been associated with an increased risk of HNC development, primarily in the head and neck region. Likewise, the interaction between systemic inflammatory disorders and immunosuppressive treatments appears to amplify the risk of HNC development, as chronic inflammation fosters a tumor-promoting microenvironment, while immunosuppressive therapies further compromise immune surveillance and anti-tumor immune responses. Understanding the molecular and cellular mechanisms underlying this interaction is crucial for developing targeted prevention strategies and therapeutic interventions. Additionally, the emerging field of immunotherapy provides potential avenues for managing HNCs associated with systemic inflammatory disorders, but further research is needed to determine its efficacy and safety in this specific context. Future studies are warranted to elucidate the underlying mechanisms and optimize preventive strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Mariana Pereira
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui Amaral Mendes
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Department of Oral and Maxillofacial Medicine and Diagnostic Sciences, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106-7401, USA
| |
Collapse
|
42
|
Yoshikawa S, Taniguchi K, Sawamura H, Ikeda Y, Asai T, Tsuji A, Matsuda S. Potential tactics with certain gut microbiota for the treatment of unresectable hepatocellular carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:556-568. [PMID: 37720344 PMCID: PMC10501893 DOI: 10.37349/etat.2023.00152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/24/2023] [Indexed: 09/19/2023] Open
Abstract
Hepatocellular carcinoma (HCC) constitutes an extremely malignant form of primary liver cancer. Intricate connections linking to the immune system might be associated with the pathogenesis of HCC. Meanwhile, immunotherapy with immune checkpoint inhibitors has been established to be a favorable therapeutic possibility for advanced HCC. Although curative opportunities for advanced HCC are restricted, the immune checkpoint immunotherapy has developed as the main choice for treating HCC. However, patients with metabolic-associated fatty liver disease (MAFLD)-linked HCC might be less likely to benefit from the immunotherapy alone. The limitation of the effect of the immunotherapy might be owing to the impaired T cell activation in MAFLD patients, which could be well explained by a dysfunctional gut-liver axis. Gut microbiota and their metabolites including several bile acids could contribute to modulating the responses of the immune checkpoint immunotherapy. Roles of gut microbiota in the development of cancers have expected great interest in the latest studies. Here, an interplay between the gut and liver has been presented, which might suggest to affect the efficacy of immune checkpoint immunotherapy against HCC.
Collapse
Affiliation(s)
- Sayuri Yoshikawa
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Kurumi Taniguchi
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Haruka Sawamura
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Yuka Ikeda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Tomoko Asai
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Ai Tsuji
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
43
|
Kayama H, Takeda K. Emerging roles of host and microbial bioactive lipids in inflammatory bowel diseases. Eur J Immunol 2023; 53:e2249866. [PMID: 37191284 DOI: 10.1002/eji.202249866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/11/2023] [Accepted: 05/15/2023] [Indexed: 05/17/2023]
Abstract
The intestinal tract harbors diverse microorganisms, host- and microbiota-derived metabolites, and potentially harmful dietary antigens. The epithelial barrier separates the mucosa, where diverse immune cells exist, from the lumen to avoid excessive immune reactions against microbes and dietary antigens. Inflammatory bowel disease (IBD), such as ulcerative colitis and Crohn's disease, is characterized by a chronic and relapsing disorder of the gastrointestinal tract. Although the precise etiology of IBD is still largely unknown, accumulating evidence suggests that IBD is multifactorial, involving host genetics and microbiota. Alterations in the metabolomic profiles and microbial community are features of IBD. Advances in mass spectrometry-based lipidomic technologies enable the identification of changes in the composition of intestinal lipid species in IBD. Because lipids have a wide range of functions, including signal transduction and cell membrane formation, the dysregulation of lipid metabolism drastically affects the physiology of the host and microorganisms. Therefore, a better understanding of the intimate interactions of intestinal lipids with host cells that are implicated in the pathogenesis of intestinal inflammation might aid in the identification of novel biomarkers and therapeutic targets for IBD. This review summarizes the current knowledge on the mechanisms by which host and microbial lipids control and maintain intestinal health and diseases.
Collapse
Affiliation(s)
- Hisako Kayama
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- WPI, Osaka University, Suita, Osaka, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, Suita, Osaka, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- WPI, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Center for Infection Disease Education and Research, Osaka University, Suita, Japan
| |
Collapse
|
44
|
Hu C, Liao S, Lv L, Li C, Mei Z. Intestinal Immune Imbalance is an Alarm in the Development of IBD. Mediators Inflamm 2023; 2023:1073984. [PMID: 37554552 PMCID: PMC10406561 DOI: 10.1155/2023/1073984] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 08/10/2023] Open
Abstract
Immune regulation plays a crucial role in human health and disease. Inflammatory bowel disease (IBD) is a chronic relapse bowel disease with an increasing incidence worldwide. Clinical treatments for IBD are limited and inefficient. However, the pathogenesis of immune-mediated IBD remains unclear. This review describes the activation of innate and adaptive immune functions by intestinal immune cells to regulate intestinal immune balance and maintain intestinal mucosal integrity. Changes in susceptible genes, autophagy, energy metabolism, and other factors interact in a complex manner with the immune system, eventually leading to intestinal immune imbalance and the onset of IBD. These events indicate that intestinal immune imbalance is an alarm for IBD development, further opening new possibilities for the unprecedented development of immunotherapy for IBD.
Collapse
Affiliation(s)
- Chunli Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shengtao Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Lin Lv
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chuanfei Li
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhechuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
45
|
Long XQ, Liu MZ, Liu ZH, Xia LZ, Lu SP, Xu XP, Wu MH. Bile acids and their receptors: Potential therapeutic targets in inflammatory bowel disease. World J Gastroenterol 2023; 29:4252-4270. [PMID: 37545642 PMCID: PMC10401658 DOI: 10.3748/wjg.v29.i27.4252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/19/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023] Open
Abstract
Chronic and recurrent inflammatory disorders of the gastrointestinal tract caused by a complex interplay between genetics and intestinal dysbiosis are called inflammatory bowel disease. As a result of the interaction between the liver and the gut microbiota, bile acids are an atypical class of steroids produced in mammals and traditionally known for their function in food absorption. With the development of genomics and metabolomics, more and more data suggest that the pathophysiological mechanisms of inflammatory bowel disease are regulated by bile acids and their receptors. Bile acids operate as signalling molecules by activating a variety of bile acid receptors that impact intestinal flora, epithelial barrier function, and intestinal immunology. Inflammatory bowel disease can be treated in new ways by using these potential molecules. This paper mainly discusses the increasing function of bile acids and their receptors in inflammatory bowel disease and their prospective therapeutic applications. In addition, we explore bile acid metabolism and the interaction of bile acids and the gut microbiota.
Collapse
Affiliation(s)
- Xiong-Quan Long
- Department of Gastroenterology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, Hunan Province, China
| | - Ming-Zhu Liu
- Department of Gastroenterology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, Hunan Province, China
| | - Zi-Hao Liu
- Department of Gastroenterology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, Hunan Province, China
| | - Lv-Zhou Xia
- Department of Gastroenterology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, Hunan Province, China
| | - Shi-Peng Lu
- Department of Gastroenterology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, Hunan Province, China
| | - Xiao-Ping Xu
- Department of Gastroenterology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, Hunan Province, China
| | - Ming-Hao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha 410005, Hunan Province, China
| |
Collapse
|
46
|
Jenkins BJ, Blagih J, Ponce-Garcia FM, Canavan M, Gudgeon N, Eastham S, Hill D, Hanlon MM, Ma EH, Bishop EL, Rees A, Cronin JG, Jury EC, Dimeloe SK, Veale DJ, Thornton CA, Vousden KH, Finlay DK, Fearon U, Jones GW, Sinclair LV, Vincent EE, Jones N. Canagliflozin impairs T cell effector function via metabolic suppression in autoimmunity. Cell Metab 2023; 35:1132-1146.e9. [PMID: 37230079 DOI: 10.1016/j.cmet.2023.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 02/03/2023] [Accepted: 05/02/2023] [Indexed: 05/27/2023]
Abstract
Augmented T cell function leading to host damage in autoimmunity is supported by metabolic dysregulation, making targeting immunometabolism an attractive therapeutic avenue. Canagliflozin, a type 2 diabetes drug, is a sodium glucose co-transporter 2 (SGLT2) inhibitor with known off-target effects on glutamate dehydrogenase and complex I. However, the effects of SGLT2 inhibitors on human T cell function have not been extensively explored. Here, we show that canagliflozin-treated T cells are compromised in their ability to activate, proliferate, and initiate effector functions. Canagliflozin inhibits T cell receptor signaling, impacting on ERK and mTORC1 activity, concomitantly associated with reduced c-Myc. Compromised c-Myc levels were encapsulated by a failure to engage translational machinery resulting in impaired metabolic protein and solute carrier production among others. Importantly, canagliflozin-treated T cells derived from patients with autoimmune disorders impaired their effector function. Taken together, our work highlights a potential therapeutic avenue for repurposing canagliflozin as an intervention for T cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Benjamin J Jenkins
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Julianna Blagih
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; University of Montreal, Maisonneuve-Rosemont Hospital Research Centre, 5414 Assomption Blvd, Montreal, QC H1T 2M4, Canada
| | - Fernando M Ponce-Garcia
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Mary Canavan
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearce Street, Dublin, Ireland
| | - Nancy Gudgeon
- Institute of Immunology and Immunotherapy, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Simon Eastham
- Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, Bristol BS8 1TD, UK
| | - David Hill
- Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, Bristol BS8 1TD, UK
| | - Megan M Hanlon
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearce Street, Dublin, Ireland
| | - Eric H Ma
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Rheos Medicines, Cambridge, MA, USA
| | - Emma L Bishop
- Institute of Immunology and Immunotherapy, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - April Rees
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - James G Cronin
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Sarah K Dimeloe
- Institute of Immunology and Immunotherapy, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Douglas J Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Catherine A Thornton
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Karen H Vousden
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - David K Finlay
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearce Street, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearce Street, Dublin, Ireland
| | - Gareth W Jones
- Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, Bristol BS8 1TD, UK
| | - Linda V Sinclair
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Emma E Vincent
- School of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Bristol BS1 3NY, UK; Integrative Epidemiology Unit, School of Population Health Science, University of Bristol, Bristol BS8 2BN, UK
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK.
| |
Collapse
|
47
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances TH17 Responses and Promotes Neutrophilic Airway Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.30.547286. [PMID: 37461622 PMCID: PMC10349957 DOI: 10.1101/2023.06.30.547286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Treatment-refractory severe asthma manifests a neutrophilic phenotype associated with TH17 responses. Heightened unfolded protein responses (UPRs) are associated with the risk of asthma, including severe asthma. However, how UPRs participate in the deregulation of TH17 cells leading to this type of asthma remains elusive. In this study, we investigated the role of the UPR sensor IRE1 in TH17 cell function and neutrophilic airway inflammation. We found that IRE1 is induced in fungal asthma and is highly expressed in TH17 cells relative to naïve CD4+ T cells. Cytokine (e.g. IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by TH17 cells. Ern1 (encoding IRE1)-deficiency decreases the expression of ER stress factors and impairs the differentiation and cytokine secretion of TH17 cells. Genetic ablation of Ern1 leads to alleviated TH17 responses and airway neutrophilia in a Candida albicans asthma model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances TH17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPRmediated secretory function of TH17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in TH17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Kourtney M. Zimmerly
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Amanda Livingston
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
48
|
Chatterjee D, Zhang Y, Ngassaki-Yoka CD, Dutilleul A, Khalfi S, Hernalsteens O, Wiche Salinas TR, Dias J, Chen H, Smail Y, Goulet JP, Bell B, Routy JP, Van Lint C, Ancuta P. Identification of aryl hydrocarbon receptor as a barrier to HIV-1 infection and outgrowth in CD4 + T cells. Cell Rep 2023; 42:112634. [PMID: 37310858 PMCID: PMC10592455 DOI: 10.1016/j.celrep.2023.112634] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 04/06/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) regulates Th17-polarized CD4+ T cell functions, but its role in HIV-1 replication/outgrowth remains unknown. Genetic (CRISPR-Cas9) and pharmacological inhibition reveal AhR as a barrier to HIV-1 replication in T cell receptor (TCR)-activated CD4+ T cells in vitro. In single-round vesicular stomatitis virus (VSV)-G-pseudotyped HIV-1 infection, AhR blockade increases the efficacy of early/late reverse transcription and subsequently facilitated integration/translation. Moreover, AhR blockade boosts viral outgrowth in CD4+ T cells of people living with HIV-1 (PLWH) receiving antiretroviral therapy (ART). Finally, RNA sequencing reveals genes/pathways downregulated by AhR blockade in CD4+ T cells of ART-treated PLWH, including HIV-1 interactors and gut-homing molecules with AhR-responsive elements in their promoters. Among them, HIC1, a repressor of Tat-mediated HIV-1 transcription and a tissue-residency master regulator, is identified by chromatin immunoprecipitation as a direct AhR target. Thus, AhR governs a T cell transcriptional program controlling viral replication/outgrowth and tissue residency/recirculation, supporting the use of AhR inhibitors in "shock and kill" HIV-1 remission/cure strategies.
Collapse
Affiliation(s)
- Debashree Chatterjee
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montréal, QC H2X 0A9, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Yuwei Zhang
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montréal, QC H2X 0A9, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Christ-Dominique Ngassaki-Yoka
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montréal, QC H2X 0A9, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Antoine Dutilleul
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université libre de Bruxelles (ULB), 6041 Gosselies, Belgium
| | - Soumia Khalfi
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Olivier Hernalsteens
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université libre de Bruxelles (ULB), 6041 Gosselies, Belgium
| | - Tomas Raul Wiche Salinas
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montréal, QC H2X 0A9, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Jonathan Dias
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montréal, QC H2X 0A9, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Huicheng Chen
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Yasmine Smail
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | | | - Brendan Bell
- Département de Microbiologie et Infectiologie, Faculté de Médecine et des Sciences de la Santé and Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Jean-Pierre Routy
- Division of Hematology and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC H3H 2R9, Canada; Infectious Disease and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC H3H 2R9, Canada
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université libre de Bruxelles (ULB), 6041 Gosselies, Belgium.
| | - Petronela Ancuta
- Centre de recherche du Centre hospitalier de l'université de Montréal, Montréal, QC H2X 0A9, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada; Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest & The Research Institute of the University of Bucharest, 050095 Bucharest, Romania.
| |
Collapse
|
49
|
Povoleri GAM, Durham LE, Gray EH, Lalnunhlimi S, Kannambath S, Pitcher MJ, Dhami P, Leeuw T, Ryan SE, Steel KJA, Kirkham BW, Taams LS. Psoriatic and rheumatoid arthritis joints differ in the composition of CD8+ tissue-resident memory T cell subsets. Cell Rep 2023; 42:112514. [PMID: 37195862 PMCID: PMC10790246 DOI: 10.1016/j.celrep.2023.112514] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/21/2023] [Accepted: 05/01/2023] [Indexed: 05/19/2023] Open
Abstract
CD69+CD103+ tissue-resident memory T (TRM) cells are important drivers of inflammation. To decipher their role in inflammatory arthritis, we apply single-cell, high-dimensional profiling to T cells from the joints of patients with psoriatic arthritis (PsA) or rheumatoid arthritis (RA). We identify three groups of synovial CD8+CD69+CD103+ TRM cells: cytotoxic and regulatory T (Treg)-like TRM cells are present in both PsA and RA, while CD161+CCR6+ type 17-like TRM cells with a pro-inflammatory cytokine profile (IL-17A+TNFα+IFNγ+) are specifically enriched in PsA. In contrast, only one population of CD4+CD69+CD103+ TRM cells is detected and at similarly low frequencies in both diseases. Type 17-like CD8+ TRM cells have a distinct transcriptomic signature and a polyclonal, but distinct, TCR repertoire. Type 17-like cells are also enriched in CD8+CD103- T cells in PsA compared with RA. These findings illustrate differences in the immunopathology of PsA and RA, with a particular enrichment for type 17 CD8+ T cells in the PsA joint.
Collapse
Affiliation(s)
- Giovanni A M Povoleri
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Lucy E Durham
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Elizabeth H Gray
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Sylvine Lalnunhlimi
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Shichina Kannambath
- BRC Genomics Core, NIHR Biomedical Research Center, Guy's and St Thomas' NHS Foundation Trust and King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Michael J Pitcher
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Pawan Dhami
- BRC Genomics Core, NIHR Biomedical Research Center, Guy's and St Thomas' NHS Foundation Trust and King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Thomas Leeuw
- Immunology & Inflammation Research TA, Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Sarah E Ryan
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Kathryn J A Steel
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Bruce W Kirkham
- Rheumatology Department, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK.
| |
Collapse
|
50
|
Wei C, Wang Y, Hu C. Bioinformatic analysis and experimental validation of the potential gene in the airway inflammation of steroid-resistant asthma. Sci Rep 2023; 13:8098. [PMID: 37208441 DOI: 10.1038/s41598-023-35214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/15/2023] [Indexed: 05/21/2023] Open
Abstract
Steroid-resistant asthma is a troublesome clinical problem in public health. The pathogenesis of steroid-resistant asthma is complex and remains to be explored. In our work, the online Gene Expression Omnibus microarray dataset GSE7368 was used to explore differentially expressed genes (DEGs) between steroid-resistant asthma patients and steroid-sensitive asthma patients. Tissue-specific gene expression of DEGs was analyzed using BioGPS. The enrichment analyses were performed using GO, KEGG, and GSEA analysis. The protein-protein interaction network and key gene cluster were constructed using STRING, Cytoscape, MCODE, and Cytohubba. A steroid-resistant neutrophilic asthma mouse model was established using lipopolysaccharide (LPS) and ovalbumin (OVA). An LPS-stimulated J744A.1 macrophage model was prepared to validate the underlying mechanism of the interesting DEG gene using the quantitative reverse transcription-polymerase chain reaction (qRT-PCR). A total of 66 DEGs were identified, most of which were present in the hematologic/immune system. Enrichment analysis displayed that the enriched pathways were the IL-17 signaling pathway, MAPK signal pathway, Toll-like receptor signaling pathway, and so on. DUSP2, as one of the top upregulated DEGs, has not been clearly demonstrated in steroid-resistant asthma. In our study, we observed that the salubrinal administration (DUSP2 inhibitor) reversed neutrophilic airway inflammation and cytokine responses (IL-17A, TNF-α) in a steroid-resistant asthma mouse model. We also found that salubrinal treatment reduced inflammatory cytokines (CXCL10 and IL-1β) in LPS-stimulated J744A.1 macrophages. DUSP2 may be a candidate target for the therapy of steroid-resistant asthma.
Collapse
Affiliation(s)
- Chaochao Wei
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Haikou, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Affiliated Hainan Hospital of Hainan Medical University, Haikou, People's Republic of China
- Department of Oncology, Xiangya Hospital Central South University, Changsha, People's Republic of China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, People's Republic of China
| | - Yang Wang
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Chengping Hu
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|