1
|
He X, Liu P, Luo Y, Fu X, Yang T. STATs, promising targets for the treatment of autoimmune and inflammatory diseases. Eur J Med Chem 2024; 277:116783. [PMID: 39180944 DOI: 10.1016/j.ejmech.2024.116783] [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: 02/05/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Cytokines play a crucial role in the pathophysiology of autoimmune and inflammatory diseases, with over 50 cytokines undergoing signal transduction through the Signal Transducers and Activators of Transcription (STAT) signaling pathway. Recent studies have solidly confirmed the pivotal role of STATs in autoimmune and inflammatory diseases. Therefore, this review provides a detailed summary of the immunological functions of STATs, focusing on exploring their mechanisms in various autoimmune and inflammatory diseases. Additionally, with the rapid advancement of structural biology in the field of drug discovery, many STAT inhibitors have been identified using structure-based drug design strategies. In this review, we also examine the structures of STAT proteins and compile the latest research on STAT inhibitors currently being tested in animal models and clinical trials for the treatment of immunological diseases, which emphasizes the feasibility of STATs as promising therapeutic targets and provides insights into the design of the next generation of STAT inhibitors.
Collapse
Affiliation(s)
- Xinlian He
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pingxian Liu
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Youfu Luo
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyuan Fu
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Yang
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Alakhras NS, Zhang W, Barros N, Sharma A, Ropa J, Priya R, Yang XF, Kaplan MH. An IL-23-STAT4 pathway is required for the proinflammatory function of classical dendritic cells during CNS inflammation. Proc Natl Acad Sci U S A 2024; 121:e2400153121. [PMID: 39088391 PMCID: PMC11317592 DOI: 10.1073/pnas.2400153121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/11/2024] [Indexed: 08/03/2024] Open
Abstract
Although many cytokine pathways are important for dendritic cell (DC) development, it is less clear what cytokine signals promote the function of mature dendritic cells. The signal transducer and activator of transcription 4 (STAT4) promotes protective immunity and autoimmunity downstream of proinflammatory cytokines including IL-12 and IL-23. In experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), Stat4-/- mice are resistant to the development of inflammation and paralysis. To define whether STAT4 is required for intrinsic signaling in mature DC function, we used conditional mutant mice in the EAE model. Deficiency of STAT4 in CD11c-expressing cells resulted in decreased T cell priming and inflammation in the central nervous system. EAE susceptibility was recovered following adoptive transfer of wild-type bone marrow-derived DCs to mice with STAT4-deficient DCs, but not adoptive transfer of STAT4- or IL-23R-deficient DCs. Single-cell RNA-sequencing (RNA-seq) identified STAT4-dependent genes in DC subsets that paralleled a signature in MS patient DCs. Together, these data define an IL-23-STAT4 pathway in DCs that is key to DC function during inflammatory disease.
Collapse
Affiliation(s)
- Nada S. Alakhras
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN46202
| | - Wenwu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN46202
| | - Nicolas Barros
- Department of Medicine, Division of Infectious Diseases Indiana University School of Medicine, Indianapolis, IN46202
| | - Anchal Sharma
- Advanced Analytics and Data Science, Eli Lilly and Company, New York, NY10016
| | - James Ropa
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN46202
| | - Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN46202
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN46202
| | - Mark H. Kaplan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN46202
| |
Collapse
|
3
|
Assiri MA, Albekairi TH, Ansari MA, Nadeem A, Attia SM, Bakheet SA, Shahid M, Aldossari AA, Almutairi MM, Almanaa TN, Alwetaid MY, Ahmad SF. The Exposure to Lead (Pb) Exacerbates Immunological Abnormalities in BTBR T + Itpr 3tf/J Mice through the Regulation of Signaling Pathways Relevant to T Cells. Int J Mol Sci 2023; 24:16218. [PMID: 38003408 PMCID: PMC10671427 DOI: 10.3390/ijms242216218] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Autism spectrum disorder (ASD) is a common neurodevelopmental illness characterized by abnormal social interactions, communication difficulties, and repetitive and limited behaviors or interests. The BTBR T+ Itpr3tf/J (BTBR) mice have been used extensively to research the ASD-like phenotype. Lead (Pb) is a hazardous chemical linked to organ damage in the human body. It is regarded as one of the most common metal exposure sources and has been connected to the development of neurological abnormalities. We used flow cytometry to investigate the molecular mechanism behind the effect of Pb exposure on subsets of CD4+ T cells in the spleen expressing IFN-γ, T-bet, STAT1, STAT4, IL-9, IRF4, IL-22, AhR, IL-10, and Foxp3. Furthermore, using RT-PCR, we studied the effect of Pb on the expression of numerous genes in brain tissue, including IFN-γ, T-bet, STAT1, STAT4, IL-9, IRF4, IL-22, AhR, IL-10, and Foxp3. Pb exposure increased the population of CD4+IFN-γ+, CD4+T-bet+, CD4+STAT1+, CD4+STAT4+, CD4+IL-9+, CD4+IRF4+, CD4+IL-22+, and CD4+AhR+ cells in BTBR mice. In contrast, CD4+IL-10+ and CD4+Foxp3+ cells were downregulated in the spleen cells of Pb-exposed BTBR mice compared to those treated with vehicle. Furthermore, Pb exposure led to a significant increase in IFN-γ, T-bet, STAT1, STAT4, IL-9, IRF4, IL-22, and AhR mRNA expression in BTBR mice. In contrast, IL-10 and Foxp3 mRNA expression was significantly lower in those treated with the vehicle. Our data suggest that Pb exposure exacerbates immunological dysfunctions associated with ASD. These data imply that Pb exposure may increase the risk of ASD.
Collapse
Affiliation(s)
- Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| | - Thamer H. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| | - Mushtaq A. Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| | - Saleh A. Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah A. Aldossari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| | - Mohammed M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| | - Taghreed N. Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia (M.Y.A.)
| | - Mohammad Y. Alwetaid
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia (M.Y.A.)
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (S.A.B.)
| |
Collapse
|
4
|
Buzzelli AA, McWilliams IL, Shin B, Bryars MT, Harrington LE. Intrinsic STAT4 Expression Controls Effector CD4 T Cell Migration and Th17 Pathogenicity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1667-1676. [PMID: 37093664 PMCID: PMC11302403 DOI: 10.4049/jimmunol.2200606] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/30/2023] [Indexed: 04/25/2023]
Abstract
Effector CD4 T cells are central to the development of autoimmune chronic inflammatory diseases, yet factors that mediate pathogenicity remain ill-defined. Single-nucleotide polymorphisms in the human STAT4 locus are associated with susceptibility to multiple autoimmune disorders, and Stat4 is linked to the pathogenic Th17 gene signature; however, Th17 cells differentiate independently of STAT4. Hence the interplay between STAT4 and CD4 T cell function, especially Th17 cells, during autoimmune disease is unclear. In this article, we demonstrate that CD4 T cell-intrinsic STAT4 expression is essential for the induction of autoimmune CNS inflammation in mice, in part by regulating the migration of CD4 T cells to the inflamed CNS. Moreover, unbiased transcriptional profiling revealed that STAT4 controls the expression of >200 genes in Th17 cells and is important for the upregulation of genes associated with IL-23-stimulated, pathogenic Th17 cells. Importantly, we show that Th17 cells specifically require STAT4 to evoke autoimmune inflammation, highlighting, to our knowledge, a novel function for STAT4 in Th17 pathogenicity.
Collapse
Affiliation(s)
- Ashlyn A. Buzzelli
- * Departments of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham. Birmingham, AL 35294
| | - Ian L. McWilliams
- * Departments of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham. Birmingham, AL 35294
| | - Boyoung Shin
- * Departments of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham. Birmingham, AL 35294
| | - Morgan T. Bryars
- * Departments of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham. Birmingham, AL 35294
| | - Laurie E. Harrington
- * Departments of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham. Birmingham, AL 35294
| |
Collapse
|
5
|
Huang J, Puente H, Wareing NE, Wu M, Mayes MD, Karmouty-Quintana H, Assassi S, Mills TW. STAT6 suppression prevents bleomycin-induced dermal fibrosis. FASEB J 2023; 37:e22761. [PMID: 36629780 PMCID: PMC10226134 DOI: 10.1096/fj.202200994r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023]
Abstract
Fibrosis of the skin and internal organs is a hallmark of systemic sclerosis (SSc). Although the pathogenesis of SSc is poorly understood, increasing evidence suggests that interleukins (IL)-4 and - 13 contribute to the pathogenesis of skin fibrosis by promoting collagen production and myofibroblast differentiation. Signal transducers and activators of transcription 6 (STAT6) is one of the most important downstream transcription factors activated by both IL-4 and IL-13. However, it is not completely understood whether STAT6 plays a role during the pathogenesis of skin fibrosis in SSc. In this study, we observed increased STAT6 phosphorylation in fibrotic skin samples collected from SSc patients as well as bleomycin-injected murine mice. Knockout of Stat6 in mice significantly (1) suppressed the expression of fibrotic cytokines including Il13, Il17, Il22, Ccl2, and the alternatively activated macrophage marker Cd206; (2) reduced the production of collagen and fibronectin, and (3) attenuated late-stage skin fibrosis and inflammation induced by bleomycin. Consistently, mice treated with STAT6 inhibitor AS1517499 also attenuated skin fibrosis on day 28. In addition, a co-culture experiment demonstrated that skin epithelial cells with STAT6 knockdown had reduced cytokine expression in response to IL-4/IL-13, and subsequently attenuated fibrotic protein expression in skin fibroblasts. On the other side, STAT6 depletion in skin fibroblasts attenuated IL-4/IL-13-induced cytokine and fibrotic marker expression, and reduced CXCL2 expression in co-cultured keratinocytes. In summary, our study highlighted an important yet not fully understood role of STAT6 in skin fibrosis by driving innate inflammation and differentiation of alternatively activated macrophages in response to injury.
Collapse
Affiliation(s)
- Jingjing Huang
- Department of Geriatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hydia Puente
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nancy E. Wareing
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Minghua Wu
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Maureen D. Mayes
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shervin Assassi
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Tingting W. Mills
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
6
|
Ahmad SF, Nadeem A, Ansari MA, Bakheet SA, Shahid M, Al-Mazroua HA, As Sobeai HM, Alasmari AF, Alanazi MM, Alhamed AS, Aldossari AA, Attia SM. CC chemokine receptor 5 antagonist alleviates inflammation by regulating IFN-γ/IL-10 and STAT4/Smad3 signaling in a mouse model of autoimmune encephalomyelitis. Cell Immunol 2022; 379:104580. [PMID: 35872534 DOI: 10.1016/j.cellimm.2022.104580] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/16/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022]
Abstract
Multiple sclerosis (MS) is an immunopathological disease that causes demyelination and recurrent episodes of T cell-mediated immune attack in the central nervous system. Experimental autoimmune encephalomyelitis (EAE) is a well-established mouse model of MS. The roles of T cells in MS/EAE have been well investigated, but little is known about the role of CCR5+ cells. In the present study, we investigated whether treatment with DAPTA, a selective CCR5 antagonist, could modulate the progression of EAE in the SJL/J mice. EAE mice were treated with DAPTA (0.01 mg/kg) intraperitoneally daily from day 14 to day 42, and the clinical scores were evaluated. We further investigated the effects of DAPTA on IFN-γ-, TGF-β-, IL-10-, IL-17A-, IL-22-, T-bet, STAT4-, RORγT-, AhR-, Smad3-, and Foxp3-expressing CCR5+ spleen cells using flow cytometry analysis. We further explored the effects of DAPTA on mRNA/protein expression of IFN-γ, IL-10, IL-17A, IL-22, TGF-β, T-bet, STAT4, RORγT, AhR, Foxp3, and NF-H in the brain tissue. The severity of clinical scores decreased in DAPTA-treated EAE mice as compared to that in the EAE control mice. Moreover, the percentage of CCR5+IFN-γ+, CCR5+T-bet+, CCR5+STAT4+, CCR5+IL-17A+, CCR5+RORγt+, CCR5+IL-22+, and CCR5+AhR+ cells decreased while CCR5+TGF-β+, CCR5+IL-10+, CCR5+Smad3+, and CCR5+Foxp3+ increased in DAPTA-treated EAE mice. Furthermore, DAPTA treatment significantly mitigated the EAE-induced expression of T-bet, STAT4, IL-17A, RORγT, IL-22, and AhR but upregulated Foxp3, IL-10, and NF-H expression in the brain tissue. Taken together, our data demonstrated that DAPTA could ameliorate EAE progression through the downregulation of the inflammation-related cytokines and transcription factors signaling, which may be useful for the clinical therapy of MS.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Homood M As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah A Aldossari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
7
|
Zehra Okus F, Busra Azizoglu Z, Canatan H, Eken A. S1P analogues SEW2871, BAF312 and FTY720 affect human Th17 and Treg generation ex vivo. Int Immunopharmacol 2022; 107:108665. [DOI: 10.1016/j.intimp.2022.108665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/05/2022]
|
8
|
Arbelaez CA, Palle P, Charaix J, Bettelli E. STAT1 signaling protects self-reactive T cells from control by innate cells during neuroinflammation. JCI Insight 2022; 7:148222. [PMID: 35587373 PMCID: PMC9309063 DOI: 10.1172/jci.insight.148222] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
The transcription factor STAT1 plays a critical role in modulating the differentiation of CD4+ T cells producing IL-17 and GM-CSF, which promote the development of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). The protective role of STAT1 in MS and EAE has been largely attributed to its ability to limit pathogenic Th cells and promote Tregs. Using mice with selective deletion of STAT1 in T cells (STAT1CD4-Cre), we identified a potentially novel mechanism by which STAT1 regulates neuroinflammation independently of Foxp3+ Tregs. STAT1-deficient effector T cells became the target of NK cell–mediated killing, limiting their capacity to induce EAE. STAT1-deficient T cells promoted their own killing by producing more IL-2 that, in return, activated NK cells. Elimination of NK cells restored EAE susceptibility in STAT1CD4-Cre mice. Therefore, our study suggests that the STAT1 pathway can be manipulated to limit autoreactive T cells during autoimmunity directed against the CNS.
Collapse
Affiliation(s)
- Carlos A Arbelaez
- Center for Fundamental Immunology, Benaroya Research Institute at Virginia Mason, Seattle, United States of America
| | - Pushpalatha Palle
- Center for Fundamental Immunology, Benaroya Research Institute at Virginia Mason, Seattle, United States of America
| | - Jonathan Charaix
- Center for Fundamental Immunology, Benaroya Research Institute at Virginia Mason, Seattle, United States of America
| | - Estelle Bettelli
- Center for Fundamental Immunology, Benaroya Research Institute at Virginia Mason, Seattle, United States of America
| |
Collapse
|
9
|
Shetty A, Tripathi SK, Junttila S, Buchacher T, Biradar R, Bhosale S, Envall T, Laiho A, Moulder R, Rasool O, Galande S, Elo L, Lahesmaa R. A systematic comparison of FOSL1, FOSL2 and BATF-mediated transcriptional regulation during early human Th17 differentiation. Nucleic Acids Res 2022; 50:4938-4958. [PMID: 35511484 PMCID: PMC9122603 DOI: 10.1093/nar/gkac256] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 03/30/2022] [Accepted: 04/19/2022] [Indexed: 12/21/2022] Open
Abstract
Th17 cells are essential for protection against extracellular pathogens, but their aberrant activity can cause autoimmunity. Molecular mechanisms that dictate Th17 cell-differentiation have been extensively studied using mouse models. However, species-specific differences underscore the need to validate these findings in human. Here, we characterized the human-specific roles of three AP-1 transcription factors, FOSL1, FOSL2 and BATF, during early stages of Th17 differentiation. Our results demonstrate that FOSL1 and FOSL2 co-repress Th17 fate-specification, whereas BATF promotes the Th17 lineage. Strikingly, FOSL1 was found to play different roles in human and mouse. Genome-wide binding analysis indicated that FOSL1, FOSL2 and BATF share occupancy over regulatory regions of genes involved in Th17 lineage commitment. These AP-1 factors also share their protein interacting partners, which suggests mechanisms for their functional interplay. Our study further reveals that the genomic binding sites of FOSL1, FOSL2 and BATF harbour hundreds of autoimmune disease-linked SNPs. We show that many of these SNPs alter the ability of these transcription factors to bind DNA. Our findings thus provide critical insights into AP-1-mediated regulation of human Th17-fate and associated pathologies.
Collapse
Affiliation(s)
| | | | | | | | - Rahul Biradar
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku 20520, Finland
| | - Santosh D Bhosale
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
- Department of Biochemistry and Molecular Biology, Protein Research Group, University of Southern Denmark, Campusvej 55, Odense M, DK 5230, Denmark
| | - Tapio Envall
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Asta Laiho
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku 20520, Finland
| | - Robert Moulder
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku 20520, Finland
| | - Omid Rasool
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku 20520, Finland
| | - Sanjeev Galande
- Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 411008, India
- Department of Life Sciences, Shiv Nadar University, Delhi-NCR
| | - Laura L Elo
- Correspondence may also be addressed to Laura Elo. Tel: +358 29 450 2090;
| | - Riitta Lahesmaa
- To whom correspondence should be addressed. Tel: +358 29 450 2415;
| |
Collapse
|
10
|
Krovi SH, Kuchroo VK. Activation pathways that drive CD4 + T cells to break tolerance in autoimmune diseases . Immunol Rev 2022; 307:161-190. [PMID: 35142369 PMCID: PMC9255211 DOI: 10.1111/imr.13071] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/11/2022]
Abstract
Autoimmune diseases are characterized by dysfunctional immune systems that misrecognize self as non-self and cause tissue destruction. Several cell types have been implicated in triggering and sustaining disease. Due to a strong association of major histocompatibility complex II (MHC-II) proteins with various autoimmune diseases, CD4+ T lymphocytes have been thoroughly investigated for their roles in dictating disease course. CD4+ T cell activation is a coordinated process that requires three distinct signals: Signal 1, which is mediated by antigen recognition on MHC-II molecules; Signal 2, which boosts signal 1 in a costimulatory manner; and Signal 3, which helps to differentiate the activated cells into functionally relevant subsets. These signals are disrupted during autoimmunity and prompt CD4+ T cells to break tolerance. Herein, we review our current understanding of how each of the three signals plays a role in three different autoimmune diseases and highlight the genetic polymorphisms that predispose individuals to autoimmunity. We also discuss the drawbacks of existing therapies and how they can be addressed to achieve lasting tolerance in patients.
Collapse
Affiliation(s)
- Sai Harsha Krovi
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
11
|
Ingelfinger F, Gerdes LA, Kavaka V, Krishnarajah S, Friebel E, Galli E, Zwicky P, Furrer R, Peukert C, Dutertre CA, Eglseer KM, Ginhoux F, Flierl-Hecht A, Kümpfel T, De Feo D, Schreiner B, Mundt S, Kerschensteiner M, Hohlfeld R, Beltrán E, Becher B. Twin study reveals non-heritable immune perturbations in multiple sclerosis. Nature 2022; 603:152-158. [PMID: 35173329 PMCID: PMC8891021 DOI: 10.1038/s41586-022-04419-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 01/04/2022] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system underpinned by partially understood genetic risk factors and environmental triggers and their undefined interactions1,2. Here we investigated the peripheral immune signatures of 61 monozygotic twin pairs discordant for MS to dissect the influence of genetic predisposition and environmental factors. Using complementary multimodal high-throughput and high-dimensional single-cell technologies in conjunction with data-driven computational tools, we identified an inflammatory shift in a monocyte cluster of twins with MS, coupled with the emergence of a population of IL-2 hyper-responsive transitional naive helper T cells as MS-related immune alterations. By integrating data on the immune profiles of healthy monozygotic and dizygotic twin pairs, we estimated the variance in CD25 expression by helper T cells displaying a naive phenotype to be largely driven by genetic and shared early environmental influences. Nonetheless, the expanding helper T cells of twins with MS, which were also elevated in non-twin patients with MS, emerged independent of the individual genetic makeup. These cells expressed central nervous system-homing receptors, exhibited a dysregulated CD25-IL-2 axis, and their proliferative capacity positively correlated with MS severity. Together, our matched-pair analysis of the extended twin approach allowed us to discern genetically and environmentally determined features of an MS-associated immune signature.
Collapse
Affiliation(s)
- Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Vladyslav Kavaka
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
| | | | - Ekaterina Friebel
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Edoardo Galli
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Neurologic Clinic and Policlinic, University Hospital Basel, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Reinhard Furrer
- Department of Mathematics, University of Zurich, Zurich, Switzerland
- Department of Computational Science, University of Zurich, Zurich, Switzerland
| | - Christian Peukert
- Department of Strategy, Globalization and Society, University of Lausanne, Lausanne, Switzerland
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Klara Magdalena Eglseer
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
| | | | - Andrea Flierl-Hecht
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Bettina Schreiner
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Martinsried, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Ansari MA, Nadeem A, Alshammari MA, Attia SM, Bakheet SA, Khan MR, Albekairi TH, Alasmari AF, Alhosaini K, Alqahtani F, Al-Mazroua HA, Ahmad SF. Cathepsin B inhibitor alleviates Th1, Th17, and Th22 transcription factor signaling dysregulation in experimental autoimmune encephalomyelitis. Exp Neurol 2022; 351:113997. [PMID: 35122866 DOI: 10.1016/j.expneurol.2022.113997] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 01/04/2022] [Accepted: 01/28/2022] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory infiltration in association with demyelination in the central nervous system. Among the factors involved in the immunological mechanisms of MS, Th1, Th17, and Th22 cells play a critical role. In the present study, we investigated the role of CA-074, a potent Cathepsin B inhibitor, in MS progression, using the SJL/J mouse model of experimental autoimmune encephalomyelitis (EAE). Following induction of EAE, mice were administered CA-074 (10 mg/kg) intraperitoneally each day, beginning on day 14 and continuing until day 28, and were evaluated for clinical signs. We further investigated the effect of CA-074 on Th1 (T-bet/STAT4), Th17 (IL-17A/RORγT), Th22 (TNF-α/IL-22), and regulatory T (Treg/Foxp3) cells in the spleen, using flow cytometry. We also analyzed the effect of CA-074 on T-bet, IL-17A, RORγT, IL-22, and mRNA and protein levels using RT-PCR and western blot analysis for brain tissues. Cathepsin B expression were also assessed by western blot in the brain tissues. The severity of clinical scores decreased significantly in CA-074-treated mice compared with that in EAE control mice. Moreover, the percentage of CD4+T-bet+, CXCR5+T-bet+, CD4+STAT4+, CD4+IL-17A+, CXCR5+IL-17A+, CD4+RORγT+, CCR6+RORγT+, CD4+TNF-α+, CD4+IL-22+, and CCR6+IL-22+ cells decreased while CD25+Foxp3+ increased in CA-074-treated EAE mice as compared to vehicle-treated EAE mice. Further, CA-074-treated EAE mice had downregulated Cathepsin B protein expression which was associated with decreased T-bet, IL-17A, RORγT, and IL-22 mRNA/protein expression. These results suggest that Cathepsin B could be a novel therapeutic candidate against for the treatment of MS.
Collapse
Affiliation(s)
- Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad R Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Thamer H Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khaled Alhosaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
13
|
Untwining Anti-Tumor and Immunosuppressive Effects of JAK Inhibitors-A Strategy for Hematological Malignancies? Cancers (Basel) 2021; 13:cancers13112611. [PMID: 34073410 PMCID: PMC8197909 DOI: 10.3390/cancers13112611] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/22/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is aberrantly activated in many malignancies. Inhibition of this pathway via JAK inhibitors (JAKinibs) is therefore an attractive therapeutic strategy underlined by Ruxolitinib (JAK1/2 inhibitor) being approved for the treatment of myeloproliferative neoplasms. As a consequence of the crucial role of the JAK-STAT pathway in the regulation of immune responses, inhibition of JAKs suppresses the immune system. This review article provides a thorough overview of the current knowledge on JAKinibs’ effects on immune cells in the context of hematological malignancies. We also discuss the potential use of JAKinibs for the treatment of diseases in which lymphocytes are the source of the malignancy. Abstract The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway propagates signals from a variety of cytokines, contributing to cellular responses in health and disease. Gain of function mutations in JAKs or STATs are associated with malignancies, with JAK2V617F being the main driver mutation in myeloproliferative neoplasms (MPN). Therefore, inhibition of this pathway is an attractive therapeutic strategy for different types of cancer. Numerous JAK inhibitors (JAKinibs) have entered clinical trials, including the JAK1/2 inhibitor Ruxolitinib approved for the treatment of MPN. Importantly, loss of function mutations in JAK-STAT members are a cause of immune suppression or deficiencies. MPN patients undergoing Ruxolitinib treatment are more susceptible to infections and secondary malignancies. This highlights the suppressive effects of JAKinibs on immune responses, which renders them successful in the treatment of autoimmune diseases but potentially detrimental for cancer patients. Here, we review the current knowledge on the effects of JAKinibs on immune cells in the context of hematological malignancies. Furthermore, we discuss the potential use of JAKinibs for the treatment of diseases in which lymphocytes are the source of malignancies. In summary, this review underlines the necessity of a robust immune profiling to provide the best benefit for JAKinib-treated patients.
Collapse
|
14
|
Pignataro G, Cataldi M, Taglialatela M. Neurological risks and benefits of cytokine-based treatments in coronavirus disease 2019: from preclinical to clinical evidence. Br J Pharmacol 2021; 179:2149-2174. [PMID: 33512003 DOI: 10.1111/bph.15397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 12/15/2022] Open
Abstract
Immunodeficiency and hyperinflammation are responsible for the most frequent and life-threatening forms of coronavirus disease 2019 (COVID-19). Therefore, cytokine-based treatments targeting immuno-inflammatory mechanisms are currently undergoing clinical scrutiny in COVID-19-affected patients. In addition, COVID-19 patients also exhibit a wide range of neurological manifestations (neuro-COVID), which may also benefit from cytokine-based treatments. In fact, such drugs have shown some clinical efficacy also in neuroinflammatory diseases. On the other hand, anti-cytokine drugs are endowed with significant neurological risks, mainly attributable to their immunodepressant effects. Therefore, the aim of the present manuscript is to briefly describe the role of specific cytokines in neuroinflammation, to summarize the efficacy in preclinical models of neuroinflammatory diseases of drugs targeting these cytokines and to review the clinical data regarding the neurological effects of these drugs currently being investigated against COVID-19, in order to raise awareness about their potentially beneficial and/or detrimental neurological consequences.
Collapse
Affiliation(s)
- Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Mauro Cataldi
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Maurizio Taglialatela
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
15
|
Effect of Cladribine on Neuronal Apoptosis: New Insight of In Vitro Study in Multiple Sclerosis Therapy. Brain Sci 2020; 10:brainsci10080548. [PMID: 32823496 PMCID: PMC7464206 DOI: 10.3390/brainsci10080548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/01/2022] Open
Abstract
Background: Cladribine (2-CdA) can cross the blood–brain barrier, resulting in inhibition of DNA synthesis and repair and disruption of cellular proliferation in actively dividing lymphocytes. No data on effect on neurons are available. Aim: To study “in vitro” 2-CdA apoptotic effects on neurons in healthy donor and multiple sclerosis patient lymphocytes. Methods: Neuroblastoma cells were co-cultured with lymphocytes, with and without 2-CdA. Results: Apoptosis increased in lymphocytes with 2-CdA; increase was also observed when lymphocytes were cultured with neuronal cells. However, neurons were not affected by 2-CdA for apoptosis. Conclusions: 2-CdA causes peripheral and central lymphocyte death preserving neurons, with a reasonable impact on inflammation and neuroprotection.
Collapse
|
16
|
Schnell A, Bod L, Madi A, Kuchroo VK. The yin and yang of co-inhibitory receptors: toward anti-tumor immunity without autoimmunity. Cell Res 2020; 30:285-299. [PMID: 31974523 PMCID: PMC7118128 DOI: 10.1038/s41422-020-0277-x] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022] Open
Abstract
Co-inhibitory receptors are important regulators of T-cell function that define the balance between tolerance and autoimmunity. The immune regulatory function of co-inhibitory receptors, including CTLA-4, PD-1, TIM-3, TIGIT, and LAG-3, was first discovered in the setting of autoimmune disease models, in which their blockade or deficiency resulted in induction or exacerbation of the disease. Later on, co-inhibitory receptors on lymphocytes have also been found to influence outcomes in tumor and chronic viral infection settings. These receptors suppress T-cell function in the tumor microenvironment (TME), thereby making the T cells dysfunctional. Based on this observation, blockade of co-inhibitory receptors (also known as checkpoint molecules) has emerged as a successful treatment option for a number of human cancers. However, severe autoimmune-like side effects limit the use of therapeutics that block individual or combinations of co-inhibitory receptors for cancer treatment. In this review we provide an overview of the role of co-inhibitory receptors in autoimmunity and anti-tumor immunity. We then discuss current approaches and future directions to leverage our knowledge of co-inhibitory receptors to target them in tumor immunity without inducing autoimmunity.
Collapse
Affiliation(s)
- Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Lloyd Bod
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
17
|
Wang X, Ni L, Wan S, Zhao X, Ding X, Dejean A, Dong C. Febrile Temperature Critically Controls the Differentiation and Pathogenicity of T Helper 17 Cells. Immunity 2020; 52:328-341.e5. [PMID: 32049050 DOI: 10.1016/j.immuni.2020.01.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/02/2019] [Accepted: 01/19/2020] [Indexed: 02/06/2023]
Abstract
Fever, an evolutionarily conserved physiological response to infection, is also commonly associated with many autoimmune diseases, but its role in T cell differentiation and autoimmunity remains largely unclear. T helper 17 (Th17) cells are critical in host defense and autoinflammatory diseases, with distinct phenotypes and pathogenicity. Here, we show that febrile temperature selectively regulated Th17 cell differentiation in vitro in enhancing interleukin-17 (IL-17), IL-17F, and IL-22 expression. Th17 cells generated under febrile temperature (38.5°C-39.5°C), compared with those under 37°C, showed enhanced pathogenic gene expression with increased pro-inflammatory activities in vivo. Mechanistically, febrile temperature promoted SUMOylation of SMAD4 transcription factor to facilitate its nuclear localization; SMAD4 deficiency selectively abrogated the effects of febrile temperature on Th17 cell differentiation both in vitro and ameliorated an autoimmune disease model. Our results thus demonstrate a critical role of fever in shaping adaptive immune responses with implications in autoimmune diseases.
Collapse
Affiliation(s)
- Xiaohu Wang
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Lu Ni
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Siyuan Wan
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaohong Zhao
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiao Ding
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Anne Dejean
- Nuclear Organization and Oncogenesis Laboratory, Department of Cell Biology and Infection, INSERM U993, Institute Pasteur, Paris 75015, France
| | - Chen Dong
- Institute of Immunology and School of Medicine, Tsinghua University, Beijing 100084, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
18
|
Wu B, Wan Y. Molecular control of pathogenic Th17 cells in autoimmune diseases. Int Immunopharmacol 2020; 80:106187. [PMID: 31931372 DOI: 10.1016/j.intimp.2020.106187] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/02/2020] [Accepted: 01/02/2020] [Indexed: 12/11/2022]
Abstract
IL-17A-producing CD4+ T helper cells (Th17) are crucial for the development of inflammatory and autoimmune diseases and thus are exploited for clinical immunotherapies. Emerging evidence suggests Th17 cells are heterogeneous and able to adopt both pathogenic and non-pathogenic phenotypes which are shaped by environmental and genetic factors. On one hand, IL-6 in concert with TGFβ1 can induce non-pathogenic Th17 cells (non-pTh17), which are not effective in inducing tissue inflammation. On the other hand, IL-6, IL-1β with IL-23 induce pathogenic Th17 cells (pTh17) to induce immune pathologies in various tissues. Th17 cells could be both pathogenic and non-pathogenic in a content-dependent manner in vivo. Understanding how the generation and pathogenicity of pTh17 cells are regulated will aid us to devise more effective immunotherapy. In this review, we summarize recent advances in the differentiation and regulation of Th17 cells especially pTh17 cells in vitro and in vivo. The emerging results revealing the specific molecular control of pTh17 cells are highlighted.
Collapse
Affiliation(s)
- Bing Wu
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Yisong Wan
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, NC 27599, USA.
| |
Collapse
|
19
|
Solouki S, August A, Huang W. Non-receptor tyrosine kinase signaling in autoimmunity and therapeutic implications. Pharmacol Ther 2019; 201:39-50. [PMID: 31082431 DOI: 10.1016/j.pharmthera.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/18/2019] [Indexed: 12/17/2022]
Abstract
Autoimmune diseases are characterized by impaired immune tolerance towards self-antigens, leading to enhanced immunity to self by dysfunctional B cells and/or T cells. The activation of these cells is controlled by non-receptor tyrosine kinases (NRTKs), which are critical mediators of antigen receptor and cytokine receptor signaling pathways. NRTKs transduce, amplify and sustain activating signals that contribute to autoimmunity, and are counter-regulated by protein tyrosine phosphatases (PTPs). The function of and interaction between NRTKs and PTPs during the development of autoimmunity could be key points of therapeutic interference against autoimmune diseases. In this review, we summarize the current state of knowledge of the functions of NRTKs and PTPs involved in B cell receptor (BCR), T cell receptor (TCR), and cytokine receptor signaling pathways that contribute to autoimmunity, and discuss their targeting for therapeutic approaches against autoimmune diseases.
Collapse
Affiliation(s)
- Sabrina Solouki
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
20
|
Zhao P, Wang P, Dong S, Zhou Z, Cao Y, Yagita H, He X, Zheng SG, Fisher SJ, Fujinami RS, Chen M. Depletion of PD-1-positive cells ameliorates autoimmune disease. Nat Biomed Eng 2019; 3:292-305. [PMID: 30952980 PMCID: PMC6452906 DOI: 10.1038/s41551-019-0360-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 01/23/2019] [Indexed: 12/11/2022]
Abstract
Targeted suppression of autoimmune diseases without collateral suppression of normal immunity remains an elusive yet clinically important goal. Targeted blockade of programmed-cell-death-protein-1 (PD-1)-an immune checkpoint factor expressed by activated T cells and B cells-is an efficacious therapy for potentiating immune activation against tumours. Here we show that an immunotoxin consisting of an anti-PD-1 single-chain variable fragment, an albumin-binding domain and Pseudomonas exotoxin targeting PD-1-expressing cells, selectively recognizes and induces the killing of the cells. Administration of the immunotoxin to mouse models of autoimmune diabetes delays disease onset, and its administration in mice paralysed by experimental autoimmune encephalomyelitis ameliorates symptoms. In all mouse models, the immunotoxin reduced the numbers of PD-1-expressing cells, of total T cells and of cells of an autoreactive T-cell clone found in inflamed organs, while maintaining active adaptive immunity, as evidenced by full-strength immune responses to vaccinations. The targeted depletion of PD-1-expressing cells contingent to the preservation of adaptive immunity might be effective in the treatment of a wide range of autoimmune diseases.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Peng Wang
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Shuyun Dong
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Zemin Zhou
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, The UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Xiao He
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Song Guo Zheng
- Division of Rheumatology, Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Simon J Fisher
- Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Mingnan Chen
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
21
|
Exploring the etiopathogenesis of systemic lupus erythematosus: a genetic perspective. Immunogenetics 2019; 71:283-297. [DOI: 10.1007/s00251-019-01103-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/07/2019] [Indexed: 12/27/2022]
|
22
|
Glatigny S, Bettelli E. Experimental Autoimmune Encephalomyelitis (EAE) as Animal Models of Multiple Sclerosis (MS). Cold Spring Harb Perspect Med 2018; 8:cshperspect.a028977. [PMID: 29311122 DOI: 10.1101/cshperspect.a028977] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a multifocal demyelinating disease of the central nervous system (CNS) leading to the progressive destruction of the myelin sheath surrounding axons. It can present with variable clinical and pathological manifestations, which might reflect the involvement of distinct pathogenic processes. Although the mechanisms leading to the development of the disease are not fully understood, numerous evidences indicate that MS is an autoimmune disease, the initiation and progression of which are dependent on an autoimmune response against myelin antigens. In addition, genetic susceptibility and environmental triggers likely contribute to the initiation of the disease. At this time, there is no cure for MS, but several disease-modifying therapies (DMTs) are available to control and slow down disease progression. A good number of these DMTs were identified and tested using animal models of MS referred to as experimental autoimmune encephalomyelitis (EAE). In this review, we will recapitulate the characteristics of EAE models and discuss how they help shed light on MS pathogenesis and help test new treatments for MS patients.
Collapse
Affiliation(s)
- Simon Glatigny
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101.,Department of Immunology, University of Washington, Seattle, Washington 98109
| | - Estelle Bettelli
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101.,Department of Immunology, University of Washington, Seattle, Washington 98109
| |
Collapse
|
23
|
Wang K, Song F, Fernandez-Escobar A, Luo G, Wang JH, Sun Y. The Properties of Cytokines in Multiple Sclerosis: Pros and Cons. Am J Med Sci 2018; 356:552-560. [PMID: 30447707 DOI: 10.1016/j.amjms.2018.08.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 08/12/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease that affects the central nervous system and is characterized by demyelination, axonal loss, gliosis and inflammation. The last plays a major role in the onset and propagation of the disease. MS presents with heterogeneous lesions containing a broad range of cells and soluble mediators of the immune system such as T cells, B cells, macrophages, microglia, cytokines, chemokines, antibodies, complement and other toxic substances. This review outlines, analyzes and discusses the different immune mechanisms of MS that are responsible for the initiation and propagation of active lesions, demyelination, axonal injury, remyelination and cell loss as well as the role of cytokines in the disease process. Proinflammatory cytokines such as interleukin-17 (IL-17), IL-22, tumor necrosis factor-α, IL-1, IL-12 and interferon-γ may cause MS through several signaling pathways. Conversely, anti-inflammatory circulating cytokines such as IL-4 and IL-10 are reduced and theoretically can exert a direct protective effect in this condition. Future studies are necessary to develop effective, safe and long-lasting strategies to reduce the abnormal cytokine cascades and to treat MS.
Collapse
Affiliation(s)
- Kexin Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Feng Song
- Qingdao University Affiliated Qingdao Municipal Hospital, Qingdao, Shandong, China
| | | | - Gang Luo
- Department of Interventional Neurology, Beijing Tiantan Hospital, Beijing, China
| | - Jun-Hui Wang
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Yu Sun
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
24
|
Xia P, Gong X, Xiao L, Wang Y, Zhang T, Liao Q, Mo X, Qiu X, Huang J. CCDC134 ameliorated experimental autoimmune encephalomyelitis by suppressing Th1 and Th17 cells. Brain Behav Immun 2018; 71:158-168. [PMID: 29548993 DOI: 10.1016/j.bbi.2018.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/22/2018] [Accepted: 03/12/2018] [Indexed: 01/12/2023] Open
Abstract
CCDC134 (coiled-coil domain containing 134), a cytokine-like molecule, was previously reported to exert antitumor effects by augmenting CD8+ T-cell mediated immunity. However, the dynamic changes in CCDC134 expression patterns in the spinal cord that may be involved in the progression of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, remains unclear. In this study, we found that CCDC134 expression was markedly increased in the spinal cord during the progression of EAE. Furthermore, we demonstrated that CCDC134 significantly reduced the severity and slowed the progression of EAE, which correlated with reduced spinal cord inflammation and demyelination. The underlying mechanism of CCDC134-induced effects involved inhibition of T helper (Th)-1 and Th17 cell differentiation and secretion of its key effector molecules IFN-γ and IL-17A via regulation of JAK/STAT signaling. These findings indicate that CCDC134 exerts potent anti-inflammatory effects through the selective modulation of pathogenic Th1 and Th17 cells by targeting critical signaling pathways. The study provides insights into the role of CCDC134 as a unique therapeutic agent for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Peng Xia
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Xiaoting Gong
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Lin Xiao
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Yida Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Tianzhuo Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Qinyuan Liao
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Xiaoning Mo
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Xiaoyan Qiu
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China
| | - Jing Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Medical Immunology, Ministry of Health, Beijing 100191, China.
| |
Collapse
|
25
|
Acharya S, Timilshina M, Jiang L, Neupane S, Choi DY, Park SW, Lee SY, Jeong BS, Kim JA, Nam TG, Chang JH. Amelioration of Experimental autoimmune encephalomyelitis and DSS induced colitis by NTG-A-009 through the inhibition of Th1 and Th17 cells differentiation. Sci Rep 2018; 8:7799. [PMID: 29773813 PMCID: PMC5958108 DOI: 10.1038/s41598-018-26088-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 05/04/2018] [Indexed: 12/13/2022] Open
Abstract
CD4+ T cells are the central for the mammalian adaptive immune system. Naïve CD4+ T cells mainly differentiate in to pro-inflammatory Th1, Th2 and Th17 cells upon antigenic stimulation. IFN-γ secreting Th1 cells and IL-17 secreting Th17 cells are found to play key roles in autoimmune diseases like multiple sclerosis (MS) and ulcerative colitis (UC). In this study we found NTG-A-009, 6-aminopyridin-3-ol, has great inhibitory effect on in vitro differentiation of Th1 and Th17 cells without affecting regulatory T cells. Moreover, NTG-A-009 had no effect on CD4+ T cell proliferation and viability. In vivo treatment has shown that NTG-A-009 has ameliorated experimental autoimmune encephalomyelitis (EAE) and dextran sulfate sodium (DSS) induced colitis through the inhibition of Th1 and Th17 cells differentiation. Mechanistically, NTG-A-009 suppressed Th1 and Th17 cells differentiation via the modulation of JAK/STAT signaling pathway. Thus, our data demonstrated that NTG-A-009 ameliorated inflammation through the inhibition of Th1 and Th17 cells generation making it a potential therapeutic candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Suman Acharya
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | | | - Liyuan Jiang
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Sabita Neupane
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Sang Won Park
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Republic of Korea
| | - Sang Yeul Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Republic of Korea
| | - Byeong-Seon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Tae-Gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Republic of Korea.
| | - Jae-Hoon Chang
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
26
|
Rotem E, Faingold O, Charni M, Klug YA, Harari D, Shmuel-Galia L, Nudelman A, Rotter V, Shai Y. The HTLV-1 gp21 fusion peptide inhibits antigen specific T-cell activation in-vitro and in mice. PLoS Pathog 2018; 14:e1007044. [PMID: 29727445 PMCID: PMC5955599 DOI: 10.1371/journal.ppat.1007044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/16/2018] [Accepted: 04/18/2018] [Indexed: 11/17/2022] Open
Abstract
The ability of the Lentivirus HIV-1 to inhibit T-cell activation by its gp41 fusion protein is well documented, yet limited data exists regarding other viral fusion proteins. HIV-1 utilizes membrane binding region of gp41 to inhibit T-cell receptor (TCR) complex activation. Here we examined whether this T-cell suppression strategy is unique to the HIV-1 gp41. We focused on T-cell modulation by the gp21 fusion peptide (FP) of the Human T-lymphotropic Virus 1 (HTLV-1), a Deltaretrovirus that like HIV infects CD4+ T-cells. Using mouse and human in-vitro T-cell models together with in-vivo T-cell hyper activation mouse model, we reveal that HTLV-1's FP inhibits T-cell activation and unlike the HIV FP, bypasses the TCR complex. HTLV FP inhibition induces a decrease in Th1 and an elevation in Th2 responses observed in mRNA, cytokine and transcription factor profiles. Administration of the HTLV FP in a T-cell hyper activation mouse model of multiple sclerosis alleviated symptoms and delayed disease onset. We further pinpointed the modulatory region within HTLV-1's FP to the same region previously identified as the HIV-1 FP active region, suggesting that through convergent evolution both viruses have obtained the ability to modulate T-cells using the same region of their fusion protein. Overall, our findings suggest that fusion protein based T-cell modulation may be a common viral trait.
Collapse
Affiliation(s)
- Etai Rotem
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Omri Faingold
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Meital Charni
- Department of molecular cell biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Yoel A Klug
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Daniel Harari
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Liraz Shmuel-Galia
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Alon Nudelman
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Varda Rotter
- Department of molecular cell biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Yechiel Shai
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
27
|
Yan Z, Gibson SA, Buckley JA, Qin H, Benveniste EN. Role of the JAK/STAT signaling pathway in regulation of innate immunity in neuroinflammatory diseases. Clin Immunol 2018; 189:4-13. [PMID: 27713030 PMCID: PMC5573639 DOI: 10.1016/j.clim.2016.09.014] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/20/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023]
Abstract
The Janus Kinase/Signal Transducers and Activators of Transcription (JAK/STAT) signaling pathway is utilized by numerous cytokines and interferons, and is essential for the development and function of both innate and adaptive immunity. Aberrant activation of the JAK/STAT pathway is evident in neuroinflammatory diseases such as Multiple Sclerosis and Parkinson's Disease. Innate immunity is the front line defender of the immune system and is composed of various cell types, including microglia, macrophages and neutrophils. Innate immune responses have both pathogenic and protective roles in neuroinflammation, depending on disease context and the microenvironment in the central nervous system. In this review, we discuss the role of innate immunity in the pathogenesis of neuroinflammatory diseases, how the JAK/STAT signaling pathway regulates the innate immune response, and finally, the potential for ameliorating neuroinflammation by utilization of JAK/STAT inhibitors.
Collapse
Affiliation(s)
- Zhaoqi Yan
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Sara A Gibson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Jessica A Buckley
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
28
|
Canto E, Isobe N, Didonna A, Hauser SL, Oksenberg JR. Aberrant STAT phosphorylation signaling in peripheral blood mononuclear cells from multiple sclerosis patients. J Neuroinflammation 2018. [PMID: 29514694 PMCID: PMC5840794 DOI: 10.1186/s12974-018-1105-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Multiple sclerosis (MS) is characterized by increased activation of peripheral blood mononuclear cells (PBMCs), linked to perturbations in the phosphorylation of signaling proteins. Methods We developed a phosphoflow cytometry protocol to assess the levels of 11 phosphorylated nuclear proteins at baseline conditions and after cell activation in distinct PBMC populations from 41 treatment-naïve relapsing-remitting (RR) MS subjects and 37 healthy controls, and in a second cohort of 9 untreated RRMS patients and 10 secondary progressive (SP) MS patients. Levels of HLA-ABC, HLA-E, and HLA-DR were also assessed. Phosphorylation levels of selected proteins were also assessed in mouse splenocytes isolated from myelin oligodendrocyte glycoprotein (MOG)35–55-induced experimental autoimmune encephalomyelitis (EAE). Results Modest differences were observed at baseline between patients and controls, with general lower phosphorylation levels in cells from affected individuals. Conversely, a dramatic increase in phosphorylated p38MAPK and STAT proteins was observed across all cell types in MS patients compared to controls after in vitro activation. A similar phosphorylation profile was observed in mouse lymphocytes primed in vivo with MOG. Furthermore, levels of all p-STAT proteins were found directly correlated with HLA expression in monocytes. Levels of phosphorylated proteins did not differ between relapsing-remitting and secondary progressive MS patients either in baseline conditions or after stimulation. Lastly, phosphorylation levels appear to be independent of the genotype. Conclusion The response to IFN-α through STAT proteins signaling is strongly dysregulated in MS patients irrespective of disease stage. These findings suggest that the aberrant activation of this pathway could lead to changes in the expression of HLA molecules in antigen presenting cells, which are known to play important roles in the regulation of the immune response in health and disease. Electronic supplementary material The online version of this article (10.1186/s12974-018-1105-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ester Canto
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Noriko Isobe
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Alessandro Didonna
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA
| | | | - Stephen L Hauser
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Jorge R Oksenberg
- Department of Neurology and Weill Institute for Neurosciences, University of California at San Francisco, Nelson Rising Lane, San Francisco, CA, 94158, USA.
| |
Collapse
|
29
|
Tanasescu R, Midgley A, Robins RA, Constantinescu CS. Decreased interferon-β induced STAT-4 activation in immune cells and clinical outcome in multiple sclerosis. Acta Neurol Scand 2017; 136:233-238. [PMID: 27918083 DOI: 10.1111/ane.12715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2016] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Interferon-β (IFN-β) is used in the treatment of multiple sclerosis (MS). IFN-β activation of signal transduction and activation of transcription (STAT)-4 is linked to its immunomodulatory effects. Previous studies suggest a type I IFN deficit in immune cells of patients MS, but data on interferon-α/β receptor (IFNAR) expression and the relationship with treatment response are conflicting. Here, we compare IFN-β-mediated STAT4 activation in immune cells of untreated patients with MS and controls. MATERIALS AND METHODS Peripheral blood mononuclear cells from 27 untreated patients with relapsing MS, obtained before the initiation of IFN-β treatment, and 12 matched controls were treated in vitro with IFN-β. Total and phosphorylated STAT4 (pSTAT4) and IFNAR were measured by flow cytometry and quantitative PCR. The patients were followed up for 5 years. RESULTS pSTAT4 induction by IFN-β was lower in patients with MS than in controls, as was expression of IFNAR. pSTAT4 expression did not correlate with the clinical outcome at 5 years, measured by EDSS change. There was a negative correlation between the baseline IFNAR1 mRNA levels and relapse rate. CONCLUSIONS The results suggest decreased IFN-β responsiveness in patients with MS, associated with reduced STAT4 activation and reduced IFNAR expression. This reduced responsiveness does not appear to affect the long-term clinical outcome of IFN-β treatment.
Collapse
Affiliation(s)
- R. Tanasescu
- Division of Clinical Neuroscience; Clinical Neurology Research Group; Queen's Medical Centre; University of Nottingham; Nottingham University Hospitals NHS Trust; Nottingham UK
- Department of Clinical Neurosciences; University of Medicine and Pharmacy Carol Davila; Department of Neurology; Colentina Hospital; Bucharest Romania
| | - A. Midgley
- Division of Clinical Neuroscience; Clinical Neurology Research Group; Queen's Medical Centre; University of Nottingham; Nottingham University Hospitals NHS Trust; Nottingham UK
| | - R. A. Robins
- Division of Immunology; Clinical Neurology Research Group; Queen's Medical Centre; University of Nottingham; Nottingham University Hospitals NHS Trust; Nottingham UK
| | - C. S. Constantinescu
- Division of Clinical Neuroscience; Clinical Neurology Research Group; Queen's Medical Centre; University of Nottingham; Nottingham University Hospitals NHS Trust; Nottingham UK
| |
Collapse
|
30
|
Zhu G, Wang X, Xiao H, Liu X, Fang Y, Zhai B, Xu R, Han G, Chen G, Hou C, Shen B, Li Y, Ma N, Wu H, Liu G, Wang R. Both Notch1 and its ligands in B cells promote antibody production. Mol Immunol 2017; 91:17-23. [PMID: 28863329 DOI: 10.1016/j.molimm.2017.08.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/21/2017] [Accepted: 08/23/2017] [Indexed: 01/01/2023]
Abstract
Notch1 signaling regulates B and T lymphocyte development and also in vitro promotes antibody secretion upon B cell activation. However, it is still unclear about the role of Notch1 in antibody production upon in vitro and in vivo mixture lymphocytes activation. We first showed that Notch1 expressed in LPS-activated CD19hi B cells and CD19cre mediated Notch1 knock-down in LPS-activated B cells. Furthermore, we demonstrated that Notch1 knock-down in B cells reduced antibody production in LPS-stimulated B cells but did not affect antibody production in LPS-stimulated splenocytes and in experimental allergic encephalomyelitis (EAE) mice. Importantly, Notch1 ligands Dll1 and Jag1 expressed in B cells and pre-coated Notch1 protein promotes Notch1-knocked down B cells to produce antibody in LPS-stimulated B cells suggesting that Notch1 in other cells may promote antibody production by binding its ligands Dll1 and Jag1 in B cells. Together, our results suggest that both Notch1 and its ligands in B cells play an important role in antibody production.
Collapse
Affiliation(s)
- Gaizhi Zhu
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China; Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, China
| | - Xiaoqian Wang
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - He Xiao
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xiaoling Liu
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China; Department of Nephrology, The 307th Hospital of Chinese People's Liberation Army, Beijing 100850, China
| | - Ying Fang
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China; Department of Rheumatology, First hospital of Jilin University, Changchun 130021, China
| | - Bing Zhai
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ruonan Xu
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Gencheng Han
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Guojiang Chen
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Chunmei Hou
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Beifen Shen
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yan Li
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ning Ma
- Department of Rheumatology, First hospital of Jilin University, Changchun 130021, China
| | - Haitao Wu
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing, China.
| | - Guangchao Liu
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, China.
| | - Renxi Wang
- Laboratory of Immunology, Institute of Basic Medical Sciences, Beijing 100850, China.
| |
Collapse
|
31
|
Ip FCF, Ng YP, Or TCT, Sun P, Fu G, Li JYH, Ye WC, Cheung TH, Ip NY. Anemoside A3 ameliorates experimental autoimmune encephalomyelitis by modulating T helper 17 cell response. PLoS One 2017; 12:e0182069. [PMID: 28759648 PMCID: PMC5536310 DOI: 10.1371/journal.pone.0182069] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
Anemoside A3 (AA3) is a natural triterpenoid glycoside isolated from the root of Pulsatilla chinensis (Bunge) Regel. We previously showed that AA3 exhibits cognitive-enhancing and neuroprotective properties. In the present study, we demonstrated that AA3 modulates inflammatory responses by regulating prostaglandin E receptor 4 signaling. Because prostaglandin E receptor 4 is involved in the pathophysiology of experimental autoimmune encephalomyelitis (EAE), an animal model of human multiple sclerosis (MS), we assessed the beneficial effect of AA3 in EAE mice. AA3 treatment significantly reduced clinical severity and inflammatory infiltrates in the spinal cord of EAE mice. In vitro studies revealed that AA3 inhibited the T cell response toward the encephalitogenic epitope of myelin oligodendrocyte glycoprotein (MOG). AA3 significantly downregulated the expressions of certain Th1 and Th17 cytokines in activated T cells re-stimulated by MOG. Moreover, AA3 inhibited the activation of STAT4 and STAT3, which are the transcription factors pivotal for Th1 and Th17 lineage differentiation, respectively, in activated T cells. Pharmacological analysis further suggested that AA3 reduced Th17 cell differentiation and expansion. In conclusion, AA3 exerts an immunomodulatory effect in EAE, demonstrating its potential as a therapeutic agent for MS in humans.
Collapse
Affiliation(s)
- Fanny C. F. Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
- HKUST–Jinan Joint Laboratory of Innovative Drug Discovery, Jinan University, Guangzhou, China
| | - Yu Pong Ng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Terry C. T. Or
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Peiran Sun
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Guangmiao Fu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Jessica Y. H. Li
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Wen-Cai Ye
- HKUST–Jinan Joint Laboratory of Innovative Drug Discovery, Jinan University, Guangzhou, China
- Institute of Traditional Chinese Medicine and Natural Products College of Pharmacy, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Tom H. Cheung
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Nancy Y. Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China
- HKUST–Jinan Joint Laboratory of Innovative Drug Discovery, Jinan University, Guangzhou, China
- * E-mail:
| |
Collapse
|
32
|
You Z, Timilshina M, Jeong BS, Chang JH. BJ-2266 ameliorates experimental autoimmune encephalomyelitis through down-regulation of the JAK/STAT signaling pathway. Eur J Immunol 2017; 47:1488-1500. [PMID: 28681958 DOI: 10.1002/eji.201646860] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 05/11/2017] [Accepted: 07/03/2017] [Indexed: 12/24/2022]
Abstract
CD4+ T cells differentiate into distinct effector subsets upon antigenic stimulation. Cytokines, and micro-environmental factors present during T-cell priming, direct differentiation of naïve CD4+ T cells into pro-inflammatory Th1 and Th17 cells. From extensive screening of 2,4,5-trimethylpyridin-3-ol derivatives with various functional groups at C(6)-position, BJ-2266, a 6-thioureido-derivative, showed potent inhibitory activity on in vitro T helper (Th)-cell differentiation. This compound inhibited IFN-γ and IL-17 production from polyclonal CD4+ T cells and ovalbumin (OVA)-specific CD4+ T cells that were activated by T-cell receptor (TCR) engagement. We assessed the inhibitory effect of BJ-2266 in experimental autoimmune encephalomyelitis (EAE). Our results suggest that BJ-2266 treatment significantly suppresses EAE disease progression with reduced generation of Th1 and Th17 cells. Notably, Th-cell differentiation was significantly suppressed by BJ-2266 treatment with no effect on apoptosis, activation and proliferation of activated T cells. Furthermore, adoptive transfer of BJ-2266 treated MOG-reactive Th1 and Th17 cells led to a lower EAE disease score and better clinical recovery from EAE. The underlying mechanism of BJ-2266 effect involved the inhibition of JAK/STAT phosphorylation that is critical for Th-cell differentiation. We conclude that BJ-2266 regulates the JAK/STAT pathway in response to cytokine signals and subsequently suppresses the differentiation of Th-cell responses.
Collapse
Affiliation(s)
- Zhiwei You
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | | | - Byeong-Seon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jae-Hoon Chang
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
33
|
Sonar SA, Shaikh S, Joshi N, Atre AN, Lal G. IFN-γ promotes transendothelial migration of CD4 + T cells across the blood-brain barrier. Immunol Cell Biol 2017; 95:843-853. [PMID: 28682305 DOI: 10.1038/icb.2017.56] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 02/08/2023]
Abstract
Transendothelial migration (TEM) of Th1 and Th17 cells across the blood-brain barrier (BBB) has a critical role in the development of experimental autoimmune encephalomyelitis (EAE). How cytokines produced by inflammatory Th1 and Th17 cells damage the endothelial BBB and promote transendothelial migration of immune cells into the central nervous system (CNS) during autoimmunity is not understood. We therefore investigated the effect of various cytokines on brain endothelial cells. Among the various cytokines tested, such as Th1 (IFN-γ, IL-1α, IL-1β, TNF-α, IL-12), Th2 (IL-3, IL-4, IL-6 and IL-13), Th17 (IL-17A, IL-17F, IL-21, IL-22, IL-23, GM-CSF) and Treg-specific cytokines (IL-10 and TGF-β), IFN-γ predominantly showed increased expression of ICAM-1, VCAM-1, MAdCAM-1, H2-Kb and I-Ab molecules on brain endothelial cells. Furthermore, IFN-γ induced transendothelial migration of CD4+ T cells from the apical (luminal side) to the basal side (abluminal side) of the endothelial monolayer to chemokine CCL21 in a STAT-1-dependent manner. IFN-γ also favored the transcellular route of TEM of CD4+ T cells. Multicolor immunofluorescence and confocal microscopic analysis showed that IFN-γ induced relocalization of ICAM-1, PECAM-1, ZO-1 and VE-cadherin in the endothelial cells, which affected the migration of CD4+ T cells. These findings reveal that the IFN-γ produced during inflammation could contribute towards disrupting the BBB and promoting TEM of CD4+ T cells. Our findings also indicate that strategies that interfere with the activation of CNS endothelial cells may help in controlling neuroinflammation and autoimmunity.
Collapse
|
34
|
Zhang J, Li Z, Hu X, Su Q, He C, Liu J, Ren H, Qian M, Liu J, Cui S, Jiang W. Knockout of P2Y 12 aggravates experimental autoimmune encephalomyelitis in mice via increasing of IL-23 production and Th17 cell differentiation by dendritic cells. Brain Behav Immun 2017; 62:245-255. [PMID: 27939246 DOI: 10.1016/j.bbi.2016.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/18/2016] [Accepted: 12/03/2016] [Indexed: 10/20/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE), a common model of multiple sclerosis (MS), is mainly mediated by CD4+ T cells with demyelination and neurodegeneration of central nervous system (CNS). The loss of P2Y12 receptor might be associated with the pathogenesis of MS/EAE, but its potential mechanism is still not clear. In this study, more severe EAE developed in P2Y12-knockout (P2Y12-KO) mice compared to WT mice. Knockout of P2Y12 increased expression of IL-17A in the sera and proportion of Th17 cells in spleen and CNS. However, in vitro studies showed that P2Y12 did not influence cell differentiation and proliferation of CD4+ T cells. In bone marrow-derived dendritic cells (BMDCs), loss of P2Y12 significantly increased the production of IL-23 in contrast to the wild-type (WT) BMDCs. FACS analysis indicated that the culture supernatant from P2Y12-deficient DCs promoted more naïve CD4+ T cells to differentiate into Th17 cells. Our finding demonstrated that genetic deletion of P2Y12 receptor broke the balance of Th subtypes by affecting the cytokine profile of BMDCs and resulted in the aggravated EAE, which suggested that P2Y12 may be a potential target in treating MS.
Collapse
Affiliation(s)
- Jiang Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhenlong Li
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xuefei Hu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Qiong Su
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Cong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jing Liu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Hua Ren
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Min Qian
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shufang Cui
- Laboratory Animal Center, Second Military Medical University, Shanghai, China.
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
35
|
Goswami R, Kaplan M. STAT Transcription Factors in T Cell Control of Health and Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:123-180. [DOI: 10.1016/bs.ircmb.2016.09.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Ni X, Geller EB, Eppihimer MJ, Eisenstein TK, Adler MW, Tuma RF. Win 55212-2, a cannabinoid receptor agonist, attenuates leukocyte/endothelial interactions in an experimental autoimmune encephalomyelitis model. Mult Scler 2016; 10:158-64. [PMID: 15124761 DOI: 10.1191/1352458504ms1009oa] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Multiple sclerosis (MS) is the most common of the immune demyelinating disorders of the central nervous system (C NS). Leukocyte/endothelial interactions are important steps in the progression of the disease and substances that interfere with these activities have been evaluated as potential therapeutic agents. C annabinoid receptor agonists have been shown to downregulate immune responses and there is preliminary evidence that they may slow the progress of MS. The purpo se of this investigation was to determine how cannabinoid recepto r agonists interfere with leukocyte rolling and adhesion. This was investigated in an experimental autoimmune encephalo myelitis (EAE) model using six to eight week old C 57BL/6 mice. Mouse myelin oligodendrocyte protein and pertussis toxin were used to induce EAE. WIN 55212-2, C B1 and C B2 antagonist were given. By use of in vivo intravital microscopy, leukocyte/endothelial interactio ns were evaluated via a cranial window implanted two days before. The results demonstrated that EAE increases leukocyte rolling and firm adhesion in the brain, and that this increased leukocyte/endothelial interactio n can be attenuated by administration of WIN 55212-2. Furthermore, use of the selective antagonists for the C B1 recepto r (SR 141716A) and the C B2 receptor (SR144528) in this study demonstrated that the cannabinoid’s inhibitory effects on leukocyte/endothelial interactions can be mediated by activating C B2 receptor.
Collapse
MESH Headings
- Animals
- Benzoxazines
- Calcium Channel Blockers/pharmacology
- Cell Adhesion/drug effects
- Cell Adhesion/immunology
- Cell Communication/drug effects
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endothelium, Vascular/cytology
- Female
- Leukocyte Rolling/drug effects
- Leukocytes/cytology
- Mice
- Mice, Inbred C57BL
- Morpholines/pharmacology
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Naphthalenes/pharmacology
- Paralysis/drug therapy
- Paralysis/immunology
- Paralysis/pathology
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB2/agonists
Collapse
Affiliation(s)
- Xiang Ni
- Department of Physiology, Temple University, 3420 N. Broad Street, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|
37
|
Ghosh D, Curtis AD, Wilkinson DS, Mannie MD. Depletion of CD4+ CD25+ regulatory T cells confers susceptibility to experimental autoimmune encephalomyelitis (EAE) in GM-CSF-deficient Csf2-/- mice. J Leukoc Biol 2016; 100:747-760. [PMID: 27256565 DOI: 10.1189/jlb.3a0815-359r] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 05/04/2016] [Indexed: 01/07/2023] Open
Abstract
Previous studies established that GM-CSF-deficient (Csf2-deficient) mice exhibit profound resistance to experimental autoimmune encephalomyelitis. This study addressed whether the resistance of Csf2-deficient mice was a result of a requirement for GM-CSF in controlling the functional balance between effector and regulatory T cell subsets during experimental autoimmune encephalomyelitis. The main observation was that treatment with the anti-CD25 mAb PC61 rendered Csf2-deficient mice fully susceptible to severe, chronic experimental autoimmune encephalomyelitis, with disease incidences and severities equivalent to that of C57BL/6 mice. When both donors and recipients were treated with PC61 in a passive model of experimental autoimmune encephalomyelitis, adoptive transfer of myelin-specific Csf2-deficient T cells into Csf2-deficient recipients resulted in a nonresolving chronic course of severe paralytic experimental autoimmune encephalomyelitis. The peripheral Csf2-deficient T cell repertoire was marked by elevated CD3+ T cell frequencies that reflected substantial accumulations of naïve CD44null-low CD4+ and CD8+ T cells but essentially normal frequencies of CD4+ CD25+ forkhead box P3+ T cells among the CD3+ T cell pool. These findings suggested that Csf2-deficient mice had secondary deficiencies in peripheral T cell sensitization to environmental antigens. In accordance, myelin oligodendrocyte glycoprotein 35-55/CFA-sensitized Csf2-deficient mice exhibited deficient peripheral sensitization to myelin oligodendrocyte glycoprotein, whereas pretreatment of Csf2-deficient mice with PC61 enabled the robust induction of myelin oligodendrocyte glycoprotein-specific T cell responses in the draining lymphatics. In conclusion, the experimental autoimmune encephalomyelitis resistance of Csf2-deficient mice, at least in part, reflects a deficient induction of effector T cell function that cannot surmount normal regulatory T cell barriers. Experimental autoimmune encephalomyelitis effector responses, however, are unleashed upon depletion of regulatory CD25+ T cells.
Collapse
Affiliation(s)
- Debjani Ghosh
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA; and
| | - Alan D Curtis
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA; and
| | - Daniel S Wilkinson
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA; and
| | - Mark D Mannie
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA; and The Harriet and John Wooten Laboratory for Alzheimer's and Neurodegenerative Disease Research, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
38
|
Goropevšek A, Holcar M, Avčin T. The Role of STAT Signaling Pathways in the Pathogenesis of Systemic Lupus Erythematosus. Clin Rev Allergy Immunol 2016; 52:164-181. [DOI: 10.1007/s12016-016-8550-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
Liu QF, Li Y, Zhao QH, Wang ZY, Hu S, Yang CQ, Ye K, Li L. Association of STAT4 rs7574865 polymorphism with susceptibility to inflammatory bowel disease: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol 2015; 39:627-36. [PMID: 26066297 DOI: 10.1016/j.clinre.2015.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/24/2015] [Accepted: 04/21/2015] [Indexed: 02/09/2023]
Abstract
OBJECTIVE Association of Signal transducers and activators of transcription-4 (STAT4) gene polymorphism with susceptibility to inflammatory bowel disease have been investigated in a number of epidemiological studies, but the results are inclusive. The aim of this meta-analysis was to more precisely estimate the relationship. METHODS The databases of Pubmed and CBM updated to October, 2014 were retrieved. Random- or fixed-effect model was used to estimate odd radio (OR) and corresponding 95% confidence interval (95%CI) on the basis of heterogeneity. RESULTS Seven articles containing 2196 Crohn's disease (CD) cases, 1588 ulcerative colitis (UC) cases and 4126 controls were identified. We detected a significant association between STAT4 rs7574865 polymorphism and IBD susceptibility in overall population (GG vs. GT+TT, OR=0.855, 95% CI=0.760-0.962, P=0.009), but not in Caucasian and Asian population, respectively. No association was detected between rs7574865 polymorphism and CD susceptibility in overall, Asian and Caucasian population, respectively. Interestingly, a significant association was detected between rs7574865 with UC susceptibility in overall population (G vs. T, OR=0.881, 95% CI=0.798-0.972, P=0.012; GG vs. GT+TT, OR=0.788, 95% CI=0.679-0.914, P=0.002; GG vs. TT, OR=0.683, 95% CI=0.498-0.937, P=0.018) and Caucasians (GG vs. GT+TT, OR=0.833, 95% CI=0.701-0.990, P=0.038; GG+GT vs. TT, OR=0.667, 95% CI=0.456-0.975, P=0.037; GG vs. TT, OR=0.636, 95% CI=0.433-0.934, P=0.021), respectively, and a possible association was found in Asian population (GG vs. GT+TT, OR=0.709, 95% CI=0.503-0.998, P=0.049). CONCLUSIONS STAT4 rs7574865 gene is IBD risk factor, and this gene polymorphism is associated with UC susceptibility, especially in Caucasians. To confirm these findings, further studies with more sample size are required for a definitive conclusion.
Collapse
Affiliation(s)
- Qi-Fei Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui Province 230601, China
| | - Yi Li
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui Province 230601, China
| | - Qi-Hong Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui Province 230601, China
| | - Zheng-Yu Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui Province 230601, China
| | - Shuang Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui Province 230601, China
| | - Chao-Qun Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui Province 230601, China
| | - Kui Ye
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui Province 230601, China
| | - Li Li
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui Province 230601, China.
| |
Collapse
|
40
|
Abstract
BACKGROUND Crohn's disease (CD) and ulcerative colitis (UC) are the major forms of inflammatory bowel disease, and pathogenesis involves a complex interplay among genetic, environmental, and immunological factors. We evaluated isoform expression of the IL-12-activated transcription factor STAT4 in children with CD and UC. METHODS We collected biopsy samples from both patients newly diagnosed with CD and with UC. We further collected blood samples from patients newly diagnosed with CD and with UC as well as from patients who had a flare-up after being in clinical remission, and we examined the ratios of STAT4β/STAT4α mRNA. In addition to STAT4 isoforms, we measured the expression of the cytokines TNFα, IFNγ, granulocyte macrophage-colony stimulating factor, and IL-17 using polymerase chain reaction of biopsy samples and multiplex analysis of patient serum samples. RESULTS Ratios of STAT4β/STAT4α were increased in specific gastrointestinal tract segments in both patients with CD and those with UC that correlate with the location and severity of inflammation. In contrast, we did not observe changes in STAT4β/STAT4α ratios in biopsy specimens from patients with eosinophilic esophagitis. We also observed increased STAT4β/STAT4α ratios in the peripheral blood mononuclear cells of patients with UC and those with CD, compared with healthy controls. Ratios were normalized after patients were treated with steroids. CONCLUSIONS Collectively, these data indicate that STAT4 isoforms could be an important noninvasive biomarker in the diagnosis and treatment of inflammatory bowel disease and that expression of these isoforms might provide further insight into the pathogenesis of IBD.
Collapse
|
41
|
Brück J, Pascolo S, Fuchs K, Kellerer C, Glocova I, Geisel J, Dengler K, Yazdi AS, Röcken M, Ghoreschi K. Cholesterol Modification of p40-Specific Small Interfering RNA Enables Therapeutic Targeting of Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:2216-23. [PMID: 26232431 DOI: 10.4049/jimmunol.1402989] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 07/01/2015] [Indexed: 11/19/2022]
Abstract
Small interfering RNA (siRNA)-based therapies allow targeted correction of molecular defects in distinct cell populations. Although efficient in multiple cell populations, dendritic cells (DCs) seem to resist siRNA delivery. Using fluorescence labeling and radiolabeling, we show that cholesterol modification enables siRNA uptake by DCs in vitro and in vivo. Delivery of cholesterol-modified p40 siRNA selectively abolished p40 transcription and suppressed TLR-triggered p40 production by DCs. During immunization with peptide in CFA, cholesterol-modified p40 siRNA generated p40-deficient, IL-10-producing DCs that prevented IL-17/Th17 and IFN-γ/Th1 responses. Only cholesterol-modified p40-siRNA established protective immunity against experimental autoimmune encephalomyelitis and suppressed IFN-γ and IL-17 expression by CNS-infiltrating mononuclear cells without inducing regulatory T cells. Because cholesterol-modified siRNA can thus modify selected DC functions in vivo, it is intriguing for targeted immune therapy of allergic, autoimmune, or neoplastic diseases.
Collapse
Affiliation(s)
- Jürgen Brück
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Steve Pascolo
- Department of Oncology, University Hospital Zurich, CH-8044 Zurich, Switzerland
| | - Kerstin Fuchs
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Christina Kellerer
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Ivana Glocova
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Julia Geisel
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Katja Dengler
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Amir S Yazdi
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| | - Kamran Ghoreschi
- Department of Dermatology, Eberhard Karls University of Tübingen, D-72076 Tübingen, Germany; and
| |
Collapse
|
42
|
STAT4 controls GM-CSF production by both Th1 and Th17 cells during EAE. J Neuroinflammation 2015; 12:128. [PMID: 26123499 PMCID: PMC4491892 DOI: 10.1186/s12974-015-0351-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/19/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, mice genetically deficient in the transcription factor signal transducer and activator of transcription 4 (STAT4) are resistant to disease. In contrast, deletion or inhibition of the Th1-associated cytokines IL-12 or IFNγ which act upstream and downstream of STAT4, respectively, does not ameliorate disease. These discordant findings imply that STAT4 may act in a non-canonical role during EAE. Recently, STAT4 has been shown to regulate GM-CSF production by CD4 T cells and this cytokine is necessary for the induction of EAE. However, it is not known if STAT4 controls GM-CSF production by both Th1 and Th17 effector CD4 T cells. METHODS This study utilized the MOG(35-55) peptide immunization model of EAE. Intracellular cytokine staining and novel mixed bone marrow chimeric mice were used to study the CD4 T cell-intrinsic role of STAT4 during disease. STAT4 chromatin-immunoprecipitation (ChIP-PCR) experiments were performed to show STAT4 directly interacts with the Csf2 gene loci. RESULTS Herein, we demonstrate that STAT4 controls CD4 T cell-intrinsic GM-CSF production by both Th1 and Th17 CD4 T cells during EAE as well as in vitro. Importantly, we show that STAT4 interacts with the Csf2 locus in MOG(35-55)-activated effector CD4 T cells demonstrating direct modulation of GM-CSF. CONCLUSIONS Overall, these studies illustrate a previously unrecognized role of STAT4 to regulate GM-CSF production by not only Th1 cells, but also Th17 effector CD4 T cell subsets during EAE pathogenesis. Critically, these data highlight for the first time that STAT4 is able to modulate the effector profile of Th17 CD4 T cell subsets, which redefines our current understanding of STAT4 as a Th1-centric factor.
Collapse
|
43
|
Boivin N, Baillargeon J, Doss PMIA, Roy AP, Rangachari M. Interferon-β suppresses murine Th1 cell function in the absence of antigen-presenting cells. PLoS One 2015; 10:e0124802. [PMID: 25885435 PMCID: PMC4401451 DOI: 10.1371/journal.pone.0124802] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/19/2015] [Indexed: 01/08/2023] Open
Abstract
Interferon (IFN)-β is a front-line therapy for the treatment of the relapsing-remitting form of multiple sclerosis. However, its immunosuppressive mechanism of function remains incompletely understood. While it has been proposed that IFN-β suppresses the function of inflammatory myelin antigen-reactive T cells by promoting the release of immunomodulatory cytokines such as IL-27 from antigen-presenting cells (APCs), its direct effects on inflammatory CD4+ Th1 cells are less clear. Here, we establish that IFN-β inhibits mouse IFN-γ+ Th1 cell function in the absence of APCs. CD4+ T cells express the type I interferon receptor, and IFN-β can suppress Th1 cell proliferation under APC-free stimulation conditions. IFN-β-treated myelin antigen-specific Th1 cells are impaired in their ability to induce severe experimental autoimmune encephalomyelitis (EAE) upon transfer to lymphocyte-deficient Rag1-/- mice. Polarized Th1 cells downregulate IFN-γ and IL-2, and upregulate the negative regulatory receptor Tim-3, when treated with IFN-β in the absence of APCs. Further, IFN-β treatment of Th1 cells upregulates phosphorylation of Stat1, and downregulates phosphorylation of Stat4. Our data indicate that IFN-γ-producing Th1 cells are directly responsive to IFN-β and point to a novel mechanism of IFN-β-mediated T cell suppression that is independent of APC-derived signals.
Collapse
Affiliation(s)
- Nicolas Boivin
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
| | - Joanie Baillargeon
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
| | - Prenitha Mercy Ignatius Arokia Doss
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Graduate Programme in Microbiology and Immunology, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
| | - Andrée-Pascale Roy
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Graduate Programme in Microbiology and Immunology, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
| | - Manu Rangachari
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
- * E-mail:
| |
Collapse
|
44
|
Benveniste EN, Liu Y, McFarland BC, Qin H. Involvement of the janus kinase/signal transducer and activator of transcription signaling pathway in multiple sclerosis and the animal model of experimental autoimmune encephalomyelitis. J Interferon Cytokine Res 2015; 34:577-88. [PMID: 25084174 DOI: 10.1089/jir.2014.0012] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) and its animal model of experimental autoimmune encephalomyelitis (EAE) are characterized by focal inflammatory infiltrates into the central nervous system, demyelinating lesions, axonal damage, and abundant production of cytokines that activate immune cells and damage neurons and oligodendrocytes, including interleukin-12 (IL-12), IL-6, IL-17, IL-21, IL-23, granulocyte macrophage-colony stimulating factor, and interferon-gamma. The Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) signaling pathway mediates the biological activities of these cytokines and is essential for the development and regulation of immune responses. Dysregulation of the JAK/STAT pathway contributes to numerous autoimmune diseases, including MS/EAE. The JAK/STAT pathway is aberrantly activated in MS/EAE because of excessive production of cytokines, loss of expression of negative regulators such as suppressors of cytokine signaling proteins, and significant enrichment of genes encoding components of the JAK/STAT pathway, including STAT3. Specific JAK/STAT inhibitors have been used in numerous preclinical models of MS and demonstrate beneficial effects on the clinical course of disease and attenuation of innate and adaptive immune responses. In addition, other drugs such as statins, glatiramer acetate, laquinimod, and fumarates have beneficial effects that involve inhibition of the JAK/STAT pathway. We conclude by discussing the feasibility of the JAK/STAT pathway as a target for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham , Birmingham, Alabama
| | | | | | | |
Collapse
|
45
|
Racke MK, Yang Y, Lovett-Racke AE. Is T-bet a potential therapeutic target in multiple sclerosis? J Interferon Cytokine Res 2015; 34:623-32. [PMID: 25084179 DOI: 10.1089/jir.2014.0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Treatments for multiple sclerosis (MS) have changed over the past years as our understanding of immunology and neuroscience has evolved. Experimental autoimmune encephalomyelitis (EAE) continues to remain the major model for MS and has been a major vehicle in the development of new therapeutic targets for MS, including new agents such as natalizumab, fingolimod, and dimethyl fumarate. As progress in the molecular understanding of immunology continues, many observations in EAE are pursued with the ultimate goal of defining the pathophysiology of MS and development of innovative treatments for the disease. Although many consider MS to be a T cell-mediated autoimmune disease directed against myelin antigens, the exact cause of the disease is still unknown. For many years, it was thought that myelin-specific T cells that secreted interferon-γ and were proinflammatory were the major T cell subset that mediated the disease, but recent studies on the cytokine phenotype of pathogenic T cells in EAE and MS have opened debate on this issue. Work over the past several years suggests that the transcription factor T-bet appears to be an important factor in T cell encephalitogenicity; however, recent data suggest that it is also dispensable in certain situations, particularly for Th17 cells. Understanding the molecular mechanisms responsible for T cell encephalitogenicity in MS and other autoimmune diseases will be essential in the development of specific therapies for these inflammatory diseases.
Collapse
Affiliation(s)
- Michael K Racke
- 1 Department of Neurology, The Ohio State University Wexner Medical Center , Columbus, Ohio
| | | | | |
Collapse
|
46
|
Abstract
INTRODUCTION STAT4, which acts as the major signaling transducing STATs in response to IL-12, is a central mediator in generating inflammation during protective immune responses and immune-mediated diseases. AREAS COVERED This review summarizes that STAT4 is essential for the differentiation and function of a wide variety of immune cells, including natural killer cells, mast cells, dendritic cells and T helper cells. In addition, STAT4-mediated signaling promoted the production of autoimmune-associated components, which are implicated in the pathogenesis of autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis and psoriasis. EXPERT OPINION Due to its crucial roles in inflammation and autoimmunity, STAT4 may have promise as an effective therapeutic target for autoimmune diseases. Understanding the molecular mechanisms driving STAT4, together with knowledge on the ability of current immunosuppressive treatment to target this process, may open an avenue to novel therapeutic options.
Collapse
Affiliation(s)
- Yan Liang
- Anhui Medical University, School of Public Health, Department of Epidemiology and Biostatistics , Anhui, PR China
| | | | | |
Collapse
|
47
|
Lin MH, Yeh LT, Chen SJ, Chiou HYC, Chu CC, Yen LB, Lin KI, Chang DM, Sytwu HK. T cell-specific BLIMP-1 deficiency exacerbates experimental autoimmune encephalomyelitis in nonobese diabetic mice by increasing Th1 and Th17 cells. Clin Immunol 2014; 151:101-13. [PMID: 24568746 DOI: 10.1016/j.clim.2014.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 02/07/2014] [Accepted: 02/08/2014] [Indexed: 01/07/2023]
Abstract
Recently, we demonstrated that B lymphocyte-induced maturation protein 1 (BLIMP-1) has a role in regulating the differentiation and effector function of Th1 and Th17 cells. As these cells play critical roles in the induction and pathogenesis of experimental autoimmune encephalomyelitis (EAE), we investigated the potential role of T cell BLIMP-1 in modulating MOG35-55-induced EAE. We established T cell-specific BLIMP-1 conditional knockout (CKO) NOD mice to dissect the role of BLIMP-1 in EAE using loss-of-function model. Our results indicate that EAE severity is dramatically exacerbated in CKO mice. The numbers of CNS-infiltrating Th1, Th17, IFN-γ(+)IL-17A(+), and IL-21(+)IL-17A(+) CD4(+) T cells are remarkably increased in brain and spinal cord of CKO mice. Moreover, the ratio of Tregs/effectors and IL-10 production of Tregs are significantly downregulated in CNS of CKO mice. We conclude that BLIMP-1 suppresses autoimmune encephalomyelitis via downregulating Th1 and Th17 cells and impairing Treg cells.
Collapse
Affiliation(s)
- Ming-Hong Lin
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Li-Tzu Yeh
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Shyi-Jou Chen
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hsin-Ying C Chiou
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chin-Chen Chu
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan, ROC; Department of Recreation and Health-Care Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan, ROC.
| | - Linju B Yen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Deh-Ming Chang
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Huey-Kang Sytwu
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan, ROC; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
48
|
Weber MS, Prod'homme T, Youssef S, Dunn SE, Steinman L, Zamvil SS. Neither T-helper type 2 nor Foxp3+ regulatory T cells are necessary for therapeutic benefit of atorvastatin in treatment of central nervous system autoimmunity. J Neuroinflammation 2014; 11:29. [PMID: 24498870 PMCID: PMC3922392 DOI: 10.1186/1742-2094-11-29] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/23/2014] [Indexed: 11/25/2022] Open
Abstract
Oral atorvastatin has prevented or reversed paralysis in the multiple sclerosis (MS) model experimental autoimmune encephalomyelitis (EAE), and reduced development of new MS lesions in clinical trials. Besides inhibiting development of encephalitogenic T cells, atorvastatin treatment of EAE has been associated with an induction of anti-inflammatory myelin-reactive T-helper type (Th)-2 cells. To investigate the clinical significance of atorvastatin-mediated Th2 differentiation, we first evaluated atorvastatin treatment in interleukin (IL)-4 green fluorescent protein-enhanced transcript (4-GET) reporter mice. Atorvastatin treatment failed to induce IL-4-producing Th2 cells in vivo; however, when T cells from atorvastatin-treated 4-GET mice were reactivated in vitro, T cells preferentially differentiated into Th2 cells, while antigen-specific T-cell proliferation and secretion of proinflammatory cytokines (interferon gamma, IL-17, tumor necrosis factor and IL-12) were reduced. Oral atorvastatin also prevented or reversed EAE in signal transducer and activator of transcription 6-deficient (STAT6−/−) mice, which cannot generate IL-4-producing Th2 cells. Further, atorvastatin treatment did not induce or expand Foxp3+ regulatory T cells in either wild-type or STAT6−/− mice. In vivo proliferation of T cells, as measured by incorporation of bromodeoxyuridine, was inhibited in atorvastatin-treated wild-type and STAT6−/− mice. These data imply that atorvastatin ameliorates central nervous system autoimmune disease primarily by inhibiting proliferation of proinflammatory encephalitogenic T cells, and not simply through induction of anti-inflammatory Th2 cells. This cytostatic effect may be a relevant mechanism of action when considering use of statins in MS and other inflammatory conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Scott S Zamvil
- Department of Neurology, University of California, 675 Nelson Rising Lane NS-215A, San Francisco, CA 94158, USA.
| |
Collapse
|
49
|
Yuan H, Feng JB, Pan HF, Qiu LX, Li LH, Zhang N, Ye DQ. A meta-analysis of the association ofSTAT4polymorphism with systemic lupus erythematosus. Mod Rheumatol 2014. [DOI: 10.3109/s10165-010-0275-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
50
|
Therapeutic modulators of STAT signalling for human diseases. Nat Rev Drug Discov 2013; 12:611-29. [PMID: 23903221 DOI: 10.1038/nrd4088] [Citation(s) in RCA: 321] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The signal transducer and activator of transcription (STAT) proteins have important roles in biological processes. The abnormal activation of STAT signalling pathways is also implicated in many human diseases, including cancer, autoimmune diseases, rheumatoid arthritis, asthma and diabetes. Over a decade has passed since the first inhibitor of a STAT protein was reported and efforts to discover modulators of STAT signalling as therapeutics continue. This Review discusses the outcomes of the ongoing drug discovery research endeavours against STAT proteins, provides perspectives on new directions for accelerating the discovery of drug candidates, and highlights the noteworthy candidate therapeutics that have progressed to clinical trials.
Collapse
|