1
|
Miao R, Liu Y, Shen S, Wang W, Wang S. Chromatin remodeling in lymphocytic function and fate: the multifaceted roles of SWI/SNF complex. Front Immunol 2025; 16:1575857. [PMID: 40342423 PMCID: PMC12058788 DOI: 10.3389/fimmu.2025.1575857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
The Switch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex comprises 10-15 subunits, which modulate the arrangement, location, or conformation of nucleosomes to upregulate chromatin accessibility. During lymphocytic differentiation and functional development, the SWI/SNF complex exerts its effects by binding to specific transcription factors (TFs) or DNA sequences via its subunits, which are thereafter recruited to the promoter or enhancer regions of target genes, rendering each subunit crucial wherein. The loss of individual subunits during lymphocytic differentiation not only disrupts the targeting of the SWI/SNF complex but also impairs its chromatin remodeling function, ultimately resulting in altered differentiation of immature lymphocytes, dysfunction of mature lymphocytes, and injured immune responses. Therefore, in this paper, we focus on TFs interacting with SWI/SNF complex subunits in lymphocytes, and summarize the effects of the loss of specific subunits of the SWI/SNF complex on lymphocytic differentiation and function, as well as the modification in the expression of key genes. We also summarize the potential clinical treatments and applications targeting the loss of SWI/SNF complex subunits, and focus on the application in Chimeric Antigen Receptor (CAR) technology. In conclusion, the SWI/SNF complex is a key regulatory factor in lymphocytic biology, involved in fundamental cellular processes and closely associated with hematological diseases and immune dysfunction. However, the specific roles of SWI/SNF complex subunits in different lymphocytic subpopulations remain unclear. Future clarification of the specific functions of these subunits in different lymphocytic subsets is expected to promote the development of immunotherapy and personalized therapy.
Collapse
Affiliation(s)
- Renjie Miao
- Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun Liu
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| | - Shuo Shen
- Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenxin Wang
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| | - Shengjun Wang
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| |
Collapse
|
2
|
Sun S, Chen Y, Ouyang Y, Tang Z. Regulatory Roles of SWI/SNF Chromatin Remodeling Complexes in Immune Response and Inflammatory Diseases. Clin Rev Allergy Immunol 2024; 68:2. [PMID: 39751934 DOI: 10.1007/s12016-024-09011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
The switch/sucrose non-fermentable (SWI/SNF) chromatin remodeling complexes (also referred to as BAF complexes) are composed of multiple subunits, which regulate the nucleosome translocation and chromatin accessibility. In recent years, significant advancements have been made in understanding mutated genes encoding subunits of the SWI/SNF complexes in cancer biology. Nevertheless, the role of SWI/SNF complexes in immune response and inflammatory diseases continues to attract significant attention. This review presents a summary of the significant functions of SWI/SNF complexes during the overall process from the development to the activation of innate and adaptive immune cells. In addition, the correlation between various SWI/SNF subunits and diverse inflammatory diseases is explored. Further investigations are warranted in terms of the mechanism of SWI/SNF complexes' preference for binding sites and opposite pro-/anti-inflammatory effects. In conclusion, further efforts are needed to evaluate the druggability of targeting SWI/SNF complexes in inflammatory diseases, and we hope this review will inspire the development of novel immune modulators in clinical practice.
Collapse
Affiliation(s)
- Shunan Sun
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuzhen Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenwei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, People's Republic of China.
| |
Collapse
|
3
|
Xia Y, Xie Y, Zhang H, Liu L. STAT4 gene polymorphisms in human diseases. Front Immunol 2024; 15:1479418. [PMID: 39575235 PMCID: PMC11578735 DOI: 10.3389/fimmu.2024.1479418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/11/2024] [Indexed: 11/24/2024] Open
Abstract
Signal transducer and activator of transcription 4 (STAT4) is a member of the STAT family, which is a group of transcription factors that regulate cytokine signaling. Genetic polymorphisms in STAT4 strongly influence immune responses and disease outcomes, especially in cancer and autoimmune diseases. Several studies have indicated that certain STAT4 gene variants are associated with alterations in STAT4 expression and/or activity and that there is a close relationship between STAT4 polymorphisms and drug efficacy. However, the underlying mechanisms are complex, and the roles of these polymorphisms in disease acquisition, progression, and severity are of widespread concern. Therefore, we provide an overview of the clinical significance of polymorphisms in STAT4 and the mechanisms by which these STAT4 variants are involved in various diseases.
Collapse
Affiliation(s)
- Yan Xia
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Minda Hospital of Hubei Minzu University, Hubei Minzu University, Enshi, Hubei, China
- Department of Nephrology, Minda Hospital Affiliated to Hubei Minzu University, Hubei Clinical Research Center for Kidney Disease, Hubei Minzu University, Enshi, Hubei, China
| | - Yanni Xie
- Department of Endocrinology, Minda Hospital Affiliated to Hubei Minzu University, Hubei Clinical Research Center for Kidney Disease, Hubei Minzu University, Enshi, Hubei, China
| | - Hao Zhang
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Minda Hospital of Hubei Minzu University, Hubei Minzu University, Enshi, Hubei, China
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Lunzhi Liu
- Department of Nephrology, Minda Hospital Affiliated to Hubei Minzu University, Hubei Clinical Research Center for Kidney Disease, Hubei Minzu University, Enshi, Hubei, China
| |
Collapse
|
4
|
Verner JM, Arbuthnott HF, Ramachandran R, Bharadwaj M, Chaudhury N, Jou E. Emerging roles of type 1 innate lymphoid cells in tumour pathogenesis and cancer immunotherapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:296-315. [PMID: 38745765 PMCID: PMC11090689 DOI: 10.37349/etat.2024.00219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/14/2023] [Indexed: 05/16/2024] Open
Abstract
Innate lymphoid cells (ILCs) are the most recently discovered class of innate immune cells found to have prominent roles in various human immune-related pathologies such as infection and autoimmune diseases. However, their role in cancer was largely unclear until recently, where several emerging studies over the past few years unanimously demonstrate ILCs to be critical players in tumour immunity. Being the innate counterpart of T cells, ILCs are potent cytokine producers through which they orchestrate the overall immune response upstream of adaptive immunity thereby modulating T cell function. Out of the major ILC subsets, ILC1s have gained significant traction as potential immunotherapeutic candidates due to their central involvement with the anti-tumour type 1 immune response. ILC1s are potent producers of the well-established anti-tumour cytokine interferon γ (IFNγ), and exert direct cytotoxicity against cancer cells in response to the cytokine interleukin-15 (IL-15). However, in advanced diseases, ILC1s are found to demonstrate an exhausted phenotype in the tumour microenvironment (TME) with impaired effector functions, characterised by decreased responsiveness to cytokines and reduced IFNγ production. Tumour cells produce immunomodulatory cytokines such as transforming growth factor β (TGFβ) and IL-23, and through these suppress ILC1 anti-tumour actfivities and converts ILC1s to pro-tumoural ILC3s respectively, resulting in disease progression. This review provides a comprehensive overview of ILC1s in tumour immunity, and discusses the exciting prospects of harnessing ILC1s for cancer immunotherapy, either alone or in combination with cytokine-based treatment. The exciting prospects of targeting the upstream innate immune system through ILC1s may surmount the limitations associated with adaptive immune T cell-based strategies used in the clinic currently, and overcome cancer immunotherapeutic resistance.
Collapse
Affiliation(s)
| | | | - Raghavskandhan Ramachandran
- Medical Sciences Division, Oxford University Hospitals, OX3 9DU Oxford, United Kingdom
- Balliol College, University of Oxford, OX1 3BJ Oxford, United Kingdom
| | - Manini Bharadwaj
- Wexham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
| | - Natasha Chaudhury
- Wexham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
| | - Eric Jou
- Medical Sciences Division, Oxford University Hospitals, OX3 9DU Oxford, United Kingdom
- Wexham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
- Kellogg College, University of Oxford, OX2 6PN Oxford, United Kingdom
| |
Collapse
|
5
|
Verner JM, Arbuthnott HF, Ramachandran R, Bharadwaj M, Chaudhury N, Jou E. Emerging roles of type 1 innate lymphoid cells in tumour pathogenesis and cancer immunotherapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:296-315. [DOI: 10.37349/etat.2023.00219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/14/2023] [Indexed: 01/04/2025] Open
Abstract
Innate lymphoid cells (ILCs) are the most recently discovered class of innate immune cells found to have prominent roles in various human immune-related pathologies such as infection and autoimmune diseases. However, their role in cancer was largely unclear until recently, where several emerging studies over the past few years unanimously demonstrate ILCs to be critical players in tumour immunity. Being the innate counterpart of T cells, ILCs are potent cytokine producers through which they orchestrate the overall immune response upstream of adaptive immunity thereby modulating T cell function. Out of the major ILC subsets, ILC1s have gained significant traction as potential immunotherapeutic candidates due to their central involvement with the anti-tumour type 1 immune response. ILC1s are potent producers of the well-established anti-tumour cytokine interferon γ (IFNγ), and exert direct cytotoxicity against cancer cells in response to the cytokine interleukin-15 (IL-15). However, in advanced diseases, ILC1s are found to demonstrate an exhausted phenotype in the tumour microenvironment (TME) with impaired effector functions, characterised by decreased responsiveness to cytokines and reduced IFNγ production. Tumour cells produce immunomodulatory cytokines such as transforming growth factor β (TGFβ) and IL-23, and through these suppress ILC1 anti-tumour actfivities and converts ILC1s to pro-tumoural ILC3s respectively, resulting in disease progression. This review provides a comprehensive overview of ILC1s in tumour immunity, and discusses the exciting prospects of harnessing ILC1s for cancer immunotherapy, either alone or in combination with cytokine-based treatment. The exciting prospects of targeting the upstream innate immune system through ILC1s may surmount the limitations associated with adaptive immune T cell-based strategies used in the clinic currently, and overcome cancer immunotherapeutic resistance.
Collapse
Affiliation(s)
| | | | - Raghavskandhan Ramachandran
- Medical Sciences Division, Oxford University Hospitals, OX3 9DU Oxford, United Kingdom; Balliol College, University of Oxford, OX1 3BJ Oxford, United Kingdom
| | - Manini Bharadwaj
- exham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
| | - Natasha Chaudhury
- exham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
| | - Eric Jou
- Medical Sciences Division, Oxford University Hospitals, OX3 9DU Oxford, United Kingdom; Wexham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom; Kellogg College, University of Oxford, OX2 6PN Oxford, United Kingdom
| |
Collapse
|
6
|
Dickerson LK, Carter JA, Kohli K, Pillarisetty VG. Emerging interleukin targets in the tumour microenvironment: implications for the treatment of gastrointestinal tumours. Gut 2023; 72:1592-1606. [PMID: 37258094 DOI: 10.1136/gutjnl-2023-329650] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
The effectiveness of antitumour immunity is dependent on intricate cytokine networks. Interleukins (ILs) are important mediators of complex interactions within the tumour microenvironment, including regulation of tumour-infiltrating lymphocyte proliferation, differentiation, migration and activation. Our evolving and increasingly nuanced understanding of the cell type-specific and heterogeneous effects of IL signalling has presented unique opportunities to fine-tune elaborate IL networks and engineer new targeted immunotherapeutics. In this review, we provide a primer for clinicians on the challenges and potential of IL-based treatment. We specifically detail the roles of IL-2, IL-10, IL-12 and IL-15 in shaping the tumour-immune landscape of gastrointestinal malignancies, paying particular attention to promising preclinical findings, early-stage clinical research and innovative therapeutic approaches that may properly place ILs to the forefront of immunotherapy regimens.
Collapse
Affiliation(s)
| | - Jason A Carter
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
| | - Karan Kohli
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
- Flatiron Bio, Palo Alto, California, USA
| | - Venu G Pillarisetty
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Kharel A, Shen J, Brown R, Chen Y, Nguyen C, Alson D, Bluemn T, Fan J, Gai K, Zhang B, Kudek M, Zhu N, Cui W. Loss of PBAF promotes expansion and effector differentiation of CD8 + T cells during chronic viral infection and cancer. Cell Rep 2023; 42:112649. [PMID: 37330910 PMCID: PMC10592487 DOI: 10.1016/j.celrep.2023.112649] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 06/20/2023] Open
Abstract
During chronic viral infection and cancer, it has been established that a subset of progenitor CD8+ T cells continuously gives rise to terminally exhausted cells and cytotoxic effector cells. Although multiple transcriptional programs governing the bifurcated differentiation trajectories have been previously studied, little is known about the chromatin structure changes regulating CD8+ T cell-fate decision. In this study, we demonstrate that the chromatin remodeling complex PBAF restrains expansion and promotes exhaustion of CD8+ T cells during chronic viral infection and cancer. Mechanistically, transcriptomic and epigenomic analyses reveal the role of PBAF in maintaining chromatin accessibility of multiple genetic pathways and transcriptional programs to restrain proliferation and promote T cell exhaustion. Harnessing this knowledge, we demonstrate that perturbation of PBAF complex constrained exhaustion and promoted expansion of tumor-specific CD8+ T cells resulting in antitumor immunity in a preclinical melanoma model, implicating PBAF as an attractive target for cancer immunotherapeutic.
Collapse
Affiliation(s)
- Arjun Kharel
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jian Shen
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ryan Brown
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yao Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Christine Nguyen
- Blood Research Institute, Versiti, Milwaukee, WI, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Donia Alson
- Blood Research Institute, Versiti, Milwaukee, WI, USA
| | - Theresa Bluemn
- Blood Research Institute, Versiti, Milwaukee, WI, USA; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jie Fan
- Department of Medicine/Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kexin Gai
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bin Zhang
- Department of Medicine/Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Microbiology-Immunology, Northwestern University, Chicago, IL, USA
| | - Matthew Kudek
- Blood Research Institute, Versiti, Milwaukee, WI, USA
| | - Nan Zhu
- Blood Research Institute, Versiti, Milwaukee, WI, USA; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Weiguo Cui
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Blood Research Institute, Versiti, Milwaukee, WI, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
8
|
Li W, Yuan J. Targeting RIPK1 kinase for modulating inflammation in human diseases. Front Immunol 2023; 14:1159743. [PMID: 36969188 PMCID: PMC10030951 DOI: 10.3389/fimmu.2023.1159743] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Receptor-Interacting Serine/Threonine-Protein Kinase 1 (RIPK1) is a master regulator of TNFR1 signaling in controlling cell death and survival. While the scaffold of RIPK1 participates in the canonical NF-κB pathway, the activation of RIPK1 kinase promotes not only necroptosis and apoptosis, but also inflammation by mediating the transcriptional induction of inflammatory cytokines. The nuclear translocation of activated RIPK1 has been shown to interact BAF-complex to promote chromatin remodeling and transcription. This review will highlight the proinflammatory role of RIPK1 kinase with focus on human neurodegenerative diseases. We will discuss the possibility of targeting RIPK1 kinase for the treatment of inflammatory pathology in human diseases.
Collapse
Affiliation(s)
- Wanjin Li
- *Correspondence: Wanjin Li, ; Junying Yuan,
| | | |
Collapse
|
9
|
Rossin F, Ciccosanti F, D'Eletto M, Occhigrossi L, Fimia GM, Piacentini M. Type 2 transglutaminase in the nucleus: the new epigenetic face of a cytoplasmic enzyme. Cell Mol Life Sci 2023; 80:52. [PMID: 36695883 PMCID: PMC9874183 DOI: 10.1007/s00018-023-04698-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/26/2023]
Abstract
One of the major mysteries in science is how it is possible to pack the cellular chromatin with a total length of over 1 m, into a small sphere with a diameter of 5 mm "the nucleus", and even more difficult to envisage how to make it functional. Although we know that compaction is achieved through the histones, however, the DNA needs to be accessible to the transcription machinery and this is allowed thanks to a variety of very complex epigenetic mechanisms. Either DNA (methylation) or post-translational modifications of histone proteins (acetylation, methylation, ubiquitination and sumoylation) play a crucial role in chromatin remodelling and consequently on gene expression. Recently the serotonylation and dopaminylation of the histone 3, catalyzed by the Transglutaminase type 2 (TG2), has been reported. These novel post-translational modifications catalyzed by a predominantly cytoplasmic enzyme opens a new avenue for future investigations on the enzyme function itself and for the possibility that other biological amines, substrate of TG2, can influence the genome regulation under peculiar cellular conditions. In this review we analyzed the nuclear TG2's biology by discussing both its post-translational modification of various transcription factors and the implications of its epigenetic new face. Finally, we will focus on the potential impact of these events in human diseases.
Collapse
Affiliation(s)
- Federica Rossin
- Department of Biology, University of Rome 'Tor Vergata', Via Della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Fabiola Ciccosanti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS 'L. Spallanzani', Rome, Italy
| | - Manuela D'Eletto
- Department of Biology, University of Rome 'Tor Vergata', Via Della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Luca Occhigrossi
- Department of Molecular Medicine, University of Rome "La Sapienza", Rome, Italy
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS 'L. Spallanzani', Rome, Italy
- Department of Molecular Medicine, University of Rome "La Sapienza", Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome 'Tor Vergata', Via Della Ricerca Scientifica 1, 00133, Rome, Italy.
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS 'L. Spallanzani', Rome, Italy.
| |
Collapse
|
10
|
Mirlekar B. Co-expression of master transcription factors determines CD4 + T cell plasticity and functions in auto-inflammatory diseases. Immunol Lett 2020; 222:58-66. [PMID: 32220615 DOI: 10.1016/j.imlet.2020.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/05/2020] [Accepted: 03/18/2020] [Indexed: 02/08/2023]
Abstract
Master CD4+ T cell lineage determined transcription factors are found to be dysregulated in pathogenesis of autoimmune and inflammatory diseases. CD4+ T cells categorized into different lineages based on their functions, cell surface markers and master transcription factors those required for expression of lineage specific cytokines. T-bet, GATA3, RORγt and Foxp3 are major transcription regulators of Th1, Th2, Th17 and Treg cells respectively. Significant progress has been made in understanding expression of lineage specific master regulators that drives CD4+ T cell differentiation. It is known that each CD4+ T cell lineage express precise determined transcription factor and due to cross regulation between these factors the CD4+ T cells able to maintain thier specific phenotype. However, recent studies shows that the lineage specifying transcription factors frequently co-expressed. There is an emerging area of research revealing that the co-expression of lineage-specifying transcription factors alters the potential function and flexibility of subsets of CD4+ T cell, this in turn favors the autoimmune pathology. Here, we discuss similarities and differences between mutually co-expressed transcription factors in CD4+ T cell subsets and then recapitulates on cell type specific and dynamic balance between the lineage restricted transcription factors in determining plasticity of CD4+ T cell subsets. Furthermore, we discuss abnormal regulation of such transcription factors that establishes a pathogenic CD4+ T cell phenotype in autoimmune diseases and how this understanding will provide further insight into potential therapeutic development.
Collapse
Affiliation(s)
- Bhalchandra Mirlekar
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, NC, 27514, USA.
| |
Collapse
|
11
|
Yang C, Mai H, Peng J, Zhou B, Hou J, Jiang D. STAT4: an immunoregulator contributing to diverse human diseases. Int J Biol Sci 2020; 16:1575-1585. [PMID: 32226303 PMCID: PMC7097918 DOI: 10.7150/ijbs.41852] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Signal transducer and activator of transcription 4 (STAT4) is a member of the STAT family and localizes to the cytoplasm. STAT4 is phosphorylated after a variety of cytokines bind to the membrane, and then dimerized STAT4 translocates to the nucleus to regulate gene expression. We reviewed the essential role played by STAT4 in a wide variety of cells and the pathogenesis of diverse human diseases, especially many kinds of autoimmune and inflammatory diseases, via activation by different cytokines through the Janus kinase (JAK)-STAT signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | - Deke Jiang
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, GuangZhou, China
| |
Collapse
|
12
|
Zhou J, Shen JY, Liu LF, Chen JS, Dou TT, Zheng M, Cai SQ. Indirect Regulation and Equilibrium of p35 and p40 Subunits of Interleukin (IL)-12/23 by Ustekinumab in Psoriasis Treatment. Med Sci Monit 2020; 26:e920371. [PMID: 31910201 PMCID: PMC6977627 DOI: 10.12659/msm.920371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Ustekinumab, a human-derived monoclonal antibody that targets the p40 subunit of interleukin (IL)-12 and IL-23, has excellent clinical efficacy and safety in treating psoriasis, with a long half-life. However, no reports have described the use of human skin/serum samples to elucidate its molecular mechanisms. MATERIAL AND METHODS Twenty-four psoriasis patients were enrolled in our double-blind study and randomly divided into placebo and ustekinumab-administered groups. Dynamic changes in psoriasis area-severity index scores, and mRNA and protein levels of p35 and p40 were analyzed at 3 time points (before treatment and during the 12th and 24th weeks of treatment). RESULTS Ustekinumab initially increased and then decreased p35 mRNA expression, but increased p40 mRNA levels throughout the study. The p35 protein levels were not significantly altered, while p40 protein levels were increased after the first 2 injections but decreased after the third injection. CONCLUSIONS We concluded that 2 equilibria influence the efficacy of ustekinumab against psoriasis. First, because of the dual roles of p35 in psoriasis pathogenesis, homeostasis occurs between p35 and p40 expression levels. The second balance lies between the upregulation of p40 mRNA levels and the ability of ustekinumab to neutralize the function of the elevated p40 protein.
Collapse
Affiliation(s)
- Jiong Zhou
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Ji-Yang Shen
- Department of Dermatology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China (mainland)
| | - Lun-Fei Liu
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland).,Department of Dermatology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China (mainland)
| | - Ji-Su Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Ting-Ting Dou
- Department of Dermatology, Shan Dong Yan Tai Nursing School, Yantai, Shandong, China (mainland)
| | - Min Zheng
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Sui-Qing Cai
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
13
|
Swiatek-Machado K, Kaminska B. STAT Signaling in Glioma Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:203-222. [PMID: 32034715 DOI: 10.1007/978-3-030-30651-9_10] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
STAT (signal transducers and activators of transcription) are latent cytoplasmic transcription factors that function as downstream effectors of cytokine and growth factor receptor signaling. The canonical JAK/STAT signaling pathway involves the activation of Janus kinases (JAK) or growth factors receptor kinases, phosphorylation of STAT proteins, their dimerization and translocation into the nucleus where STATs act as transcription factors with pleiotropic downstream effects. STAT signaling is tightly controlled with restricted kinetics due to action of its negative regulators. While STAT1 is believed to play an important role in growth arrest and apoptosis, and to act as a tumor suppressor, STAT3 and 5 are involved in promoting cell cycle progression, cellular transformation, and preventing apoptosis. Aberrant activation of STATs, in particular STAT3 and STAT5, have been found in a large number of human tumors, including gliomas and may contribute to oncogenesis. In this chapter, we have (1) summarized the mechanisms of STAT activation in normal and malignant signaling; (2) discussed evidence for the critical role of constitutively activated STAT3 and STAT5 in glioma pathobiology; (3) disclosed molecular and pharmacological strategies to interfere with STAT signaling for potential therapeutic intervention in gliomas.
Collapse
Affiliation(s)
- Karolina Swiatek-Machado
- Laboratory of Transcription Regulation, Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St, PL 02-093, Warsaw, Poland.
| | - Bozena Kaminska
- Laboratory of Transcription Regulation, Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St, PL 02-093, Warsaw, Poland
| |
Collapse
|
14
|
Martin MD, Jensen IJ, Ishizuka AS, Lefebvre M, Shan Q, Xue HH, Harty JT, Seder RA, Badovinac VP. Bystander responses impact accurate detection of murine and human antigen-specific CD8 T cells. J Clin Invest 2019; 129:3894-3908. [PMID: 31219804 DOI: 10.1172/jci124443] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Induction of memory CD8 T cells is important for controlling infections such as malaria HIV/AIDS, and for cancer immunotherapy. Accurate assessment of antigen (Ag)-specific CD8 T-cells is critical for vaccine optimization and defining correlates of protection. However, conditions for determining Ag-specific CD8 T-cell responses ex-vivo using ICS may be variable, especially in humans with complex antigens. Here, we used an attenuated whole parasite malaria vaccine model in humans and various experimental infections in mice to show that the duration of antigenic stimulation and timing of brefeldin A (BFA) addition influences the magnitude of Ag-specific and bystander T cell responses. Indeed, following immunization with an attenuated whole sporozoite malaria vaccine in humans, significantly higher numbers of IFN-γ producing memory CD8 T-cells comprised of antigen specific and bystander responses were detected by increasing the duration of Ag-stimulation prior to addition of BFA. Mechanistic analyses of virus-specific CD8 T-cells in mice revealed that the increase in IFNg producing CD8 T-cells was due to bystander activation of Ag-experienced memory CD8 T-cells, and correlated with the proportion of Ag-experienced CD8 T-cells in the stimulated populations. Incubation with anti-cytokine antibodies (ex. IL-12) improved accuracy in detecting bona-fide memory CD8 T-cell responses suggesting this as the mechanism for the bystander activation. These data have important implications for accurate assessment of immune responses generated by vaccines intended to elicit protective memory CD8 T-cells.
Collapse
Affiliation(s)
| | - Isaac J Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Andrew S Ishizuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Mitchell Lefebvre
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Qiang Shan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Hai-Hui Xue
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
| | - John T Harty
- Department of Pathology and.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Vladimir P Badovinac
- Department of Pathology and.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
15
|
Bianchi E, Rogge L. The IL-23/IL-17 pathway in human chronic inflammatory diseases – new insight from genetics and targeted therapies. Microbes Infect 2019; 21:246-253. [DOI: 10.1016/j.micinf.2019.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
|
16
|
The IL-23/IL-17 pathway in human chronic inflammatory diseases-new insight from genetics and targeted therapies. Genes Immun 2019; 20:415-425. [PMID: 31000797 DOI: 10.1038/s41435-019-0067-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/15/2022]
Abstract
Chronic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, spondyloarthritis, and psoriasis cause significant morbidity and are a considerable burden for the patients in terms of pain, impaired function, and diminished quality of life, as well as for society, because of the associated high health-care costs and loss of productivity. Our limited understanding of the pathogenic mechanisms involved in these diseases currently hinders early diagnosis and the development of more specific and effective therapies. The past years have been marked by considerable progress in our insight of the genetic basis of many diseases. In particular, genome-wide association studies (GWAS) performed with thousands of patients have provided detailed information about the genetic variants associated with a large number of chronic inflammatory diseases. These studies have brought to the forefront many genes linked to signaling pathways that were not previously known to be involved in pathogenesis, pointing to new directions in the study of disease mechanisms. GWAS also provided fundamental evidence for a key role of the immune system in the pathogenesis of these diseases, because many of the identified loci map to genes involved in different immune processes. However, the mechanisms by which disease-associated genetic variants act on disease development and the targeted cell populations remain poorly understood. The challenge of the post-GWAS era is to understand how these variants affect pathogenesis, to allow translation of genetic data into better diagnostics and innovative treatment strategies. Here, we review recent results that document the importance of the IL-23/IL-17 pathway for the pathogenesis of several chronic inflammatory diseases and summarize data that demonstrate how therapeutic targeting of this pathway can benefit affected patients.
Collapse
|
17
|
Yin S, Yu J, Hu B, Lu C, Liu X, Gao X, Li W, Zhou L, Wang J, Wang D, Lu L, Wang L. Runx3 Mediates Resistance to Intracellular Bacterial Infection by Promoting IL12 Signaling in Group 1 ILC and NCR+ILC3. Front Immunol 2018; 9:2101. [PMID: 30258450 PMCID: PMC6144956 DOI: 10.3389/fimmu.2018.02101] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/24/2018] [Indexed: 12/20/2022] Open
Abstract
Innate lymphoid cells (ILCs) are the most recently identified family of the innate immune system and are hypothesized to modulate immune functions prior to the generation of adaptive immune responses. Subsets of ILCs reside in the mucosa and regulate immune responses to external pathogens; however, their role and the mechanism by which they protect against intracellular bacterial infection is not completely understood. In this report, using S. typhimurium and L. monocytogenes, we found that the levels of group 1 ILCs and NCR+ ILC3s were increased upon infection and that these increases were associated with Runt-related transcription factor 3 (Runx3) expression. Runx3 fl/fl PLZF-cre mice were much more sensitive to infection with the intracellular bacterial pathogens S. typhimurium and L. monocytogenes partially due to abnormal Group 1 ILC and NCR+ILC3 function. We also found that Runx3 directly binds to the Il12Rβ2 promoter and intron 8 to accelerate the expression of Il12Rβ2 and modulates IFNγ secretion triggered by the IL12/ STAT4 axis. Therefore, we demonstrate that Runx3 influences group 1 ILC- and NCR+ILC3-mediated immune protection against intracellular bacterial infections of both the gut and liver.
Collapse
Affiliation(s)
- Shengxia Yin
- Institute of Immunology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Jingjing Yu
- Institute of Immunology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Bian Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chenyu Lu
- Institute of Immunology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Xia Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xianzhi Gao
- Institute of Immunology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Wei Li
- Laboraty Animal Center, Zhejiang University, Hangzhou, China
| | - Lina Zhou
- Institute of Immunology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Jianli Wang
- Institute of Immunology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Di Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Linrong Lu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lie Wang
- Institute of Immunology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China.,Laboraty Animal Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Fang P, Li X, Dai J, Cole L, Camacho JA, Zhang Y, Ji Y, Wang J, Yang XF, Wang H. Immune cell subset differentiation and tissue inflammation. J Hematol Oncol 2018; 11:97. [PMID: 30064449 PMCID: PMC6069866 DOI: 10.1186/s13045-018-0637-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023] Open
Abstract
Immune cells were traditionally considered as major pro-inflammatory contributors. Recent advances in molecular immunology prove that immune cell lineages are composed of different subsets capable of a vast array of specialized functions. These immune cell subsets share distinct duties in regulating innate and adaptive immune functions and contribute to both immune activation and immune suppression responses in peripheral tissue. Here, we summarized current understanding of the different subsets of major immune cells, including T cells, B cells, dendritic cells, monocytes, and macrophages. We highlighted molecular characterization, frequency, and tissue distribution of these immune cell subsets in human and mice. In addition, we described specific cytokine production, molecular signaling, biological functions, and tissue population changes of these immune cell subsets in both cardiovascular diseases and cancers. Finally, we presented a working model of the differentiation of inflammatory mononuclear cells, their interaction with endothelial cells, and their contribution to tissue inflammation. In summary, this review offers an updated and comprehensive guideline for immune cell development and subset differentiation, including subset characterization, signaling, modulation, and disease associations. We propose that immune cell subset differentiation and its complex interaction within the internal biological milieu compose a “pathophysiological network,” an interactive cross-talking complex, which plays a critical role in the development of inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Pu Fang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jin Dai
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Lauren Cole
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Javier Andres Camacho
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Yuling Zhang
- Cardiovascular Medicine Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Jingfeng Wang
- Cardiovascular Medicine Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.,Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA. .,Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Rapp M, Lau CM, Adams NM, Weizman OE, O'Sullivan TE, Geary CD, Sun JC. Core-binding factor β and Runx transcription factors promote adaptive natural killer cell responses. Sci Immunol 2018; 2:2/18/eaan3796. [PMID: 29222089 DOI: 10.1126/sciimmunol.aan3796] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/12/2017] [Accepted: 11/02/2017] [Indexed: 01/03/2023]
Abstract
Natural killer (NK) cells are innate lymphocytes that have features of adaptive immunity such as clonal expansion and generation of long-lived memory. Interleukin-12 (IL-12) signaling through its downstream transcription factor signal transducer and activator of transcription 4 (STAT4) is required for the generation of memory NK cells after expansion. We identify gene loci that are highly enriched for STAT4 binding using chromatin immunoprecipitation sequencing for STAT4 and the permissive histone mark H3K4me3 in activated NK cells. We found that promoter regions of Runx1 and Runx3 are targets of STAT4 and that STAT4 binding during NK cell activation induces epigenetic modifications of Runx gene loci resulting in increased expression. Furthermore, specific ablation of Runx1, Runx3, or their binding partner Cbfb in NK cells resulted in defective clonal expansion and memory formation during viral infection, with evidence for Runx1-mediated control of a cell cycle program. Thus, our study reveals a mechanism whereby STAT4-mediated epigenetic control of individual Runx transcription factors promotes the adaptive behavior of antiviral NK cells.
Collapse
Affiliation(s)
- Moritz Rapp
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Colleen M Lau
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nicholas M Adams
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Orr-El Weizman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Timothy E O'Sullivan
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Clair D Geary
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
20
|
Genes uniquely expressed in human growth plate chondrocytes uncover a distinct regulatory network. BMC Genomics 2017; 18:983. [PMID: 29262782 PMCID: PMC5738906 DOI: 10.1186/s12864-017-4378-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 12/11/2017] [Indexed: 01/05/2023] Open
Abstract
Background Chondrogenesis is the earliest stage of skeletal development and is a highly dynamic process, integrating the activities and functions of transcription factors, cell signaling molecules and extracellular matrix proteins. The molecular mechanisms underlying chondrogenesis have been extensively studied and multiple key regulators of this process have been identified. However, a genome-wide overview of the gene regulatory network in chondrogenesis has not been achieved. Results In this study, employing RNA sequencing, we identified 332 protein coding genes and 34 long non-coding RNA (lncRNA) genes that are highly selectively expressed in human fetal growth plate chondrocytes. Among the protein coding genes, 32 genes were associated with 62 distinct human skeletal disorders and 153 genes were associated with skeletal defects in knockout mice, confirming their essential roles in skeletal formation. These gene products formed a comprehensive physical interaction network and participated in multiple cellular processes regulating skeletal development. The data also revealed 34 transcription factors and 11,334 distal enhancers that were uniquely active in chondrocytes, functioning as transcriptional regulators for the cartilage-selective genes. Conclusions Our findings revealed a complex gene regulatory network controlling skeletal development whereby transcription factors, enhancers and lncRNAs participate in chondrogenesis by transcriptional regulation of key genes. Additionally, the cartilage-selective genes represent candidate genes for unsolved human skeletal disorders. Electronic supplementary material The online version of this article (10.1186/s12864-017-4378-y) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Gu J, Crosier PS, Hall CJ, Chen L, Xu X. Inflammatory pathway network-based drug repositioning and molecular phenomics. MOLECULAR BIOSYSTEMS 2016; 12:2777-84. [PMID: 27345454 DOI: 10.1039/c6mb00222f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Inflammation is a protective biological response to body/tissue damage that involves immune cells, blood vessels and molecular mediators. In this work, we constructed the pathway network of inflammation, including 11 sub-pathways of inflammatory factors. Pathway-based network efficiency and network flux were adopted to evaluate drug efficacy. By using approved and experimentally validated anti-inflammatory drugs as training sets, a predictive model was built to screen potential anti-inflammatory drugs from approved drugs in DrugBank. This drug repositioning approach would bring a fast and cheap way to find new indications for approved drugs. Moreover, molecular phenomics profiles of the expression of inflammatory factors will provide new insight into the drug mechanism of action.
Collapse
Affiliation(s)
- Jiangyong Gu
- Beijing National Laboratory for Molecular Sciences, State Key Lab of Rare Earth Material Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - Christopher J Hall
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand.
| | - Lirong Chen
- Beijing National Laboratory for Molecular Sciences, State Key Lab of Rare Earth Material Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Xiaojie Xu
- Beijing National Laboratory for Molecular Sciences, State Key Lab of Rare Earth Material Chemistry and Applications, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| |
Collapse
|
22
|
Sun L, Wu Q, Han B, Li G, Sun Z, Zhang J, An L. Mechanisms of immune injury and heterogeneity of bone marrow hematopoietic cells island in patients with auto-immuno-related hematocytopenia. J Immunoassay Immunochem 2015; 35:378-87. [PMID: 24666376 DOI: 10.1080/15321819.2014.899251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Because of environmental pollution more and more people are suffered with auto-immuno-related hematocytopenia (AIRH). Serum IL-12, IL-17, and IFN-γ levels were detected by ELISA and lymphocyte subsets were analyzed by flow cytometry. Peroxidase (POX) and HLA-DR of immune cells were detected by cytochemical and immunochemical staining. Cells expressing anti-human IgG, FcγR II, MR, and other molecules in HI were detected by immunofluorescence. Serum IL-12, IL-17, and IFN-γ levels of patients were significantly higher than control group. Lymphocyte subsets of patients showed that the percentages of CD19+ B cells and CD3+ CD8+ T cell in peripheral blood were both significantly elevated. HI were mainly classified into three types, in these three types of hematopoietic cells island, peroxidase, and HLA-DR expression varied. Hematopoietic cells with pathological changes expressed anti-human IgG. The immunocytes with different levels of immunomolecules adhered captured and devoured abnormal hematopoietic cells. Immune cells expressed IL-12, IL-17A, and IL-17RA, leading to inflammatory injury of hematopoietic cells. HI destroys cells which connect auto-antibodies. Immune cells in HI express a variety of immune molecules, promote cell immune responses, and amplify the inflammatory reaction by ADCC effect or phagocytosis. These ultimately destruct directly and damage indirectly hematopoietic cells.
Collapse
Affiliation(s)
- LiFei Sun
- a Department of Tumor Research and Treatment Centricity , People's Liberation Army No. 148 Hospital , Zibo , Shandong Province , China
| | | | | | | | | | | | | |
Collapse
|
23
|
Disruption of hSWI/SNF complexes in T cells by WAS mutations distinguishes X-linked thrombocytopenia from Wiskott-Aldrich syndrome. Blood 2014; 124:3409-19. [PMID: 25253772 DOI: 10.1182/blood-2014-07-587642] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Wiskott-Aldrich syndrome (WAS), an immunodeficiency disorder, and X-linked thrombocytopenia (XLT), a bleeding disorder, both arise from nonsynonymous mutations in WAS, which encodes a hematopoietic-specific WASp. Intriguingly, XLT evolves into WAS in some patients but not in others; yet the biological basis for this cross-phenotype (CP) effect remains unclear. Using human T-helper (TH) cells expressing different disease-causing WAS mutations, we demonstrated that hSWI/SNF-like complexes require nuclear-WASp to execute their chromatin-remodeling activity at promoters of WASp-target, immune function genes during TH1 differentiation. Hot-spot WAS mutations Thr45Met and Arg86Cys, which result in XLT-to-WAS disease progression, impair recruitment of hBRM- but not BRG1-enriched BAF complexes to IFNG and TBX21 promoters. Moreover, promoter enrichment of histone H2A.Z and its catalyzing enzyme EP400 are both impaired. Consequently, activation of Notch signaling, a hBRM-regulated event, and its downstream effector NF-κB are both compromised, along with decreased accessibility of nucleosomal DNA and inefficient transcription-elongation of WASp-target TH1 genes. In contrast, patient mutations Ala236Gly and Arg477Lys that manifest in XLT without progressing to WAS do not disrupt chromatin remodeling or transcriptional reprogramming of TH1 genes. Our study defines an indispensable relationship between nuclear-WASp- and hSWI/SNF-complexes in gene activation and reveals molecular distinctions in TH cells that might contribute to disease severity in the XLT/WAS clinical spectrum.
Collapse
|
24
|
Shih HY, Sciumè G, Poholek AC, Vahedi G, Hirahara K, Villarino AV, Bonelli M, Bosselut R, Kanno Y, Muljo SA, O'Shea JJ. Transcriptional and epigenetic networks of helper T and innate lymphoid cells. Immunol Rev 2014; 261:23-49. [PMID: 25123275 PMCID: PMC4321863 DOI: 10.1111/imr.12208] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of the specification of CD4(+) helper T cells to discrete effector 'lineages' represented a watershed event in conceptualizing mechanisms of host defense and immunoregulation. However, our appreciation for the actual complexity of helper T-cell subsets continues unabated. Just as the Sami language of Scandinavia has 1000 different words for reindeer, immunologists recognize the range of fates available for a CD4(+) T cell is numerous and may be underestimated. Added to the crowded scene for helper T-cell subsets is the continuously growing family of innate lymphoid cells (ILCs), endowed with common effector responses and the previously defined 'master regulators' for CD4(+) helper T-cell subsets are also shared by ILC subsets. Within the context of this extraordinary complexity are concomitant advances in the understanding of transcriptomes and epigenomes. So what do terms like 'lineage commitment' and helper T-cell 'specification' mean in the early 21st century? How do we put all of this together in a coherent conceptual framework? It would be arrogant to assume that we have a sophisticated enough understanding to seriously answer these questions. Instead, we review the current status of the flexibility of helper T-cell responses in relation to their genetic regulatory networks and epigenetic landscapes. Recent data have provided major surprises as to what master regulators can or cannot do, how they interact with other transcription factors and impact global genome-wide changes, and how all these factors come together to influence helper cell function.
Collapse
Affiliation(s)
- Han-Yu Shih
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kyläniemi MK, Kaukonen R, Myllyviita J, Rasool O, Lahesmaa R. The regulation and role of c-FLIP in human Th cell differentiation. PLoS One 2014; 9:e102022. [PMID: 25019384 PMCID: PMC4096760 DOI: 10.1371/journal.pone.0102022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 06/14/2014] [Indexed: 12/19/2022] Open
Abstract
The early differentiation of T helper (Th) cells is a tightly controlled and finely balanced process, which involves several factors including cytokines, transcription factors and co-stimulatory molecules. Recent studies have shown that in addition to the regulation of apoptosis, caspase activity is also needed for Th cell proliferation and activation and it might play a role in Th cell differentiation. The isoforms of the cellular FLICE inhibitory protein (c-FLIP) are regulators of CASPASE-8 activity and the short isoform, c-FLIPS, has been shown to be up-regulated by IL-4, the Th2 driving cytokine. In this work, we have studied the expression and functional role of three c-FLIP isoforms during the early Th cell differentiation. Only two of the isoforms, c-FLIPS and c-FLIPL, were detected at the protein level although c-FLIPR was expressed at the mRNA level. The knockdown of c-FLIPL led to enhanced Th1 differentiation and elevated IL-4 production by Th2 cells, whereas the knockdown of c-FLIPS diminished GATA3 expression and IL-4 production by Th2 cells. In summary, our results provide new insight into the role of c-FLIP proteins in the early differentiation of human Th cells.
Collapse
Affiliation(s)
- Minna K. Kyläniemi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- National Doctoral Programme in Informational and Structural Biology, Åbo Akademi University, Turku, Finland
| | - Riina Kaukonen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Myllyviita
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Omid Rasool
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- * E-mail:
| |
Collapse
|
26
|
Cichocki F, Schlums H, Li H, Stache V, Holmes T, Lenvik TR, Chiang SCC, Miller JS, Meeths M, Anderson SK, Bryceson YT. Transcriptional regulation of Munc13-4 expression in cytotoxic lymphocytes is disrupted by an intronic mutation associated with a primary immunodeficiency. J Exp Med 2014; 211:1079-91. [PMID: 24842371 PMCID: PMC4042637 DOI: 10.1084/jem.20131131] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 04/11/2014] [Indexed: 11/16/2022] Open
Abstract
Autosomal recessive mutations in UNC13D, the gene that encodes Munc13-4, are associated with familial hemophagocytic lymphohistiocytosis type 3 (FHL3). Munc13-4 expression is obligatory for exocytosis of lytic granules, facilitating cytotoxicity by T cells and natural killer (NK) cells. The mechanisms regulating Munc13-4 expression are unknown. Here, we report that Munc13-4 is highly expressed in differentiated human NK cells and effector CD8(+) T lymphocytes. A UNC13D c.118-308C>T mutation, causative of FHL3, disrupted binding of the ETS family member ELF1 to a conserved intronic sequence. This mutation impairs UNC13D intron 1 recruitment of STAT4 and the chromatin remodeling complex component BRG1, diminishing active histone modifications at the locus. The intronic sequence acted as an overall enhancer of Munc13-4 expression in cytotoxic lymphocytes in addition to representing an alternative promoter encoding a novel Munc13-4 isoform. Mechanistically, T cell receptor engagement facilitated STAT4-dependent Munc13-4 expression in naive CD8(+) T lymphocytes. Collectively, our data demonstrates how chromatin remodeling within an evolutionarily conserved regulatory element in intron 1 of UNC13D regulates the induction of Munc13-4 expression in cytotoxic lymphocytes and suggests that an alternative Munc13-4 isoform is required for lymphocyte cytotoxicity. Thus, mutations associated with primary immunodeficiencies may cause disease by disrupting transcription factor binding.
Collapse
Affiliation(s)
- Frank Cichocki
- Centre for Infectious Medicine, Department of Medicine; Clinical Genetics Unit, Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden Division of Hematology, Oncology and Transplantation, University of Minnesota Cancer Center, Minneapolis, MN 55455
| | - Heinrich Schlums
- Centre for Infectious Medicine, Department of Medicine; Clinical Genetics Unit, Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Hongchuan Li
- Basic Science Program, Leidos Biomedical Research, Inc., Laboratory of Experimental Immunology, SAIC-Frederick Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Vanessa Stache
- Centre for Infectious Medicine, Department of Medicine; Clinical Genetics Unit, Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Timothy Holmes
- Centre for Infectious Medicine, Department of Medicine; Clinical Genetics Unit, Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Todd R Lenvik
- Division of Hematology, Oncology and Transplantation, University of Minnesota Cancer Center, Minneapolis, MN 55455
| | - Samuel C C Chiang
- Centre for Infectious Medicine, Department of Medicine; Clinical Genetics Unit, Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Jeffrey S Miller
- Division of Hematology, Oncology and Transplantation, University of Minnesota Cancer Center, Minneapolis, MN 55455
| | - Marie Meeths
- Centre for Infectious Medicine, Department of Medicine; Clinical Genetics Unit, Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Stephen K Anderson
- Basic Science Program, Leidos Biomedical Research, Inc., Laboratory of Experimental Immunology, SAIC-Frederick Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Yenan T Bryceson
- Centre for Infectious Medicine, Department of Medicine; Clinical Genetics Unit, Department of Molecular Medicine and Surgery, and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden Broegelmann Research Laboratory, Clinical Institute, University of Bergen, N-5021 Bergen, Norway
| |
Collapse
|
27
|
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
|
28
|
Abstract
In higher eukaryotic organisms epigenetic modifications are crucial for proper chromatin folding and thereby proper regulation of gene expression. In the last years the involvement of aberrant epigenetic modifications in inflammatory and autoimmune diseases has been recognized and attracted significant interest. However, the epigenetic mechanisms underlying the different disease phenotypes are still poorly understood. As autoimmune and inflammatory diseases are at least partly T cell mediated, we will provide in this chapter an introduction to the epigenetics of T cell differentiation followed by a summary of the current knowledge on aberrant epigenetic modifications that dysfunctional T cells display in various diseases such as type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, and asthma.
Collapse
|
29
|
Coffre M, Roumier M, Rybczynska M, Sechet E, Law HKW, Gossec L, Dougados M, Bianchi E, Rogge L. Combinatorial control of Th17 and Th1 cell functions by genetic variations in genes associated with the interleukin-23 signaling pathway in spondyloarthritis. ACTA ACUST UNITED AC 2013; 65:1510-21. [PMID: 23508476 DOI: 10.1002/art.37936] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 03/07/2013] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Recent genome-wide association studies have revealed numerous genetic associations between specific single-nucleotide polymorphisms (SNPs) and immune-mediated inflammatory diseases. The current challenge is to identify associations of the genetic variants with effector mechanisms implicated in pathogenesis. This study was undertaken to investigate the link between genetic variation at loci associated with spondyloarthritis (SpA) and the effector function of CD4+ T lymphocyte subsets involved in chronic inflammatory disease. METHODS Expression of Th17 and Th1 cytokines and transcription factors was measured in CD4+ T cells isolated from patients with SpA. Correlation analyses were performed to assess potential associations of these expression levels with the patient's genotype at loci genetically linked to SpA. RESULTS The effector functions of Th17 and Th1 cells in patients with SpA were found to be under combinatorial control by multiple SNPs at genes associated with the interleukin-23 (IL-23)/Th17 pathway. Patients with SpA carrying risk-associated alleles of genes in the IL-23/Th17 pathway expressed the highest levels of genes involved in the differentiation and function of Th17 and Th1 cells, whereas the presence of protective alleles was associated with low-level expression of these genes. In contrast, variation at loci that were genetically linked to SpA, but not associated with the IL-23 pathway, did not affect the expression of Th17- and Th1-specific genes, suggesting that these SNPs may contribute to the pathogenesis of SpA through distinct cellular mechanisms. CONCLUSION These results show that genetic variations at genes associated with the IL-23 signaling pathway may influence the effector functions of Th17 and Th1 cells in patients with SpA. These findings provide a framework to delineate the mechanisms by which genetic variants contribute to pathology.
Collapse
|
30
|
Corre B, Perrier J, El Khouri M, Cerboni S, Pellegrini S, Michel F. Type I interferon potentiates T-cell receptor mediated induction of IL-10-producing CD4⁺ T cells. Eur J Immunol 2013; 43:2730-40. [PMID: 23839924 DOI: 10.1002/eji.201242977] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 04/26/2013] [Accepted: 06/18/2013] [Indexed: 12/24/2022]
Abstract
Type I interferons (IFNs) have the dual ability to promote the development of the immune response and exert an anti-inflammatory activity. We analyzed the integrated effect of IFN-α, TCR signal strength, and CD28 costimulation on human CD4⁺ T-cell differentiation into cell subsets producing the anti- and proinflammatory cytokines IL-10 and IFN-γ. We show that IFN-α boosted TCR-induced IL-10 expression in activated peripheral CD45RA⁺CD4⁺ T cells and in whole blood cultures. The functional cooperation between TCR and IFN-α efficiently occurred at low engagement of receptors. Moreover, IFN-α rapidly cooperated with anti-CD3 stimulation alone. IFN-α, but not IL-10, drove the early development of type I regulatory T cells that were mostly IL-10⁺ Foxp3⁻ IFN-γ⁻ and favored IL-10 expression in a fraction of Foxp3⁺ T cells. Our data support a model in which IFN-α costimulates TCR toward the production of IL-10 whose level can be amplified via an autocrine feedback loop.
Collapse
Affiliation(s)
- Béatrice Corre
- Department of Immunology, Unit of Cytokine Signaling, Institut Pasteur, Paris, France
| | | | | | | | | | | |
Collapse
|
31
|
Pham D, Yu Q, Walline CC, Muthukrishnan R, Blum JS, Kaplan MH. Opposing roles of STAT4 and Dnmt3a in Th1 gene regulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:902-11. [PMID: 23772023 PMCID: PMC3703830 DOI: 10.4049/jimmunol.1203229] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The STAT transcription factor STAT4 is a critical regulator of Th1 differentiation and inflammatory disease. Yet, how STAT4 regulates gene expression is still unclear. In this report, we define a STAT4-dependent sequence of events including histone H3 lysine 4 methylation, Jmjd3 association with STAT4 target loci, and a Jmjd3-dependent decrease in histone H3 lysine 27 trimethylation and DNA methyltransferase (Dnmt) 3a association with STAT4 target loci. Dnmt3a has an obligate role in repressing Th1 gene expression, and in Th1 cultures deficient in both STAT4 and Dnmt3a, there is recovery in the expression of a subset of Th1 genes that is sufficient to increase IFN-γ production. Moreover, although STAT4-deficient mice are protected from the development of experimental autoimmune encephalomyelitis, mice deficient in STAT4 and conditionally deficient in Dnmt3a in T cells develop paralysis. Th1 genes that are derepressed in the absence of Dnmt3a have greater induction after the ectopic expression of the Th1-associated transcription factors T-bet and Hlx1. Together, these data demonstrate that STAT4 and Dnmt3a play opposing roles in regulating Th1 gene expression, and that one mechanism for STAT4-dependent gene programming is in establishing a derepressed genetic state susceptible to transactivation by additional fate-determining transcription factors.
Collapse
Affiliation(s)
- Duy Pham
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | |
Collapse
|
32
|
Shaposhnikov AV, Komar’kov IF, Lebedeva LA, Shidlovskii YV. Molecular components of JAK/STAT signaling pathway and its interaction with transcription machinery. Mol Biol 2013. [DOI: 10.1134/s0026893313030126] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Dong J, Chang HD, Ivascu C, Qian Y, Rezai S, Okhrimenko A, Cosmi L, Maggi L, Eckhardt F, Wu P, Sieper J, Alexander T, Annunziato F, Gossen M, Li J, Radbruch A, Thiel A. Loss of methylation at the IFNG promoter and CNS-1 is associated with the development of functional IFN-γ memory in human CD4(+) T lymphocytes. Eur J Immunol 2013; 43:793-804. [PMID: 23255246 DOI: 10.1002/eji.201242858] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 11/19/2012] [Accepted: 12/12/2012] [Indexed: 12/17/2022]
Abstract
Cytokine memory for IFN-γ production by effector/memory Th1 cells plays a key role in both protective and pathological immune responses. To understand the epigenetic mechanism determining the ontogeny of effector/memory Th1 cells characterized by stable effector functions, we identified a T-cell-specific methylation pattern at the IFNG promoter and CNS-1 in ex vivo effector/memory Th1 cells, and investigated methylation dynamics of these regions during the development of effector/memory Th1 cells. During Th1 differentiation, demethylation occurred at both the promoter and CNS-1 regions of IFNG as early as 16 h, and this process was independent of cell proliferation and DNA synthesis. Using an IFN-γ capture assay, we found early IFN-γ-producing cells from 2-day differentiating cultures acquired "permissive" levels of demethylation and developed into effector/memory Th1 cells undergoing progressive demethylation at the IFNG promoter and CNS-1 when induced by IL-12. Methylation levels of these regions in effector/memory Th1 cells of peripheral blood from rheumatoid arthritis patients correlated inversely with reduced frequencies of IFN-γ-producers, coincident with recruitment of effector/memory Th1 cells to the site of inflammation. Thus, after termination of TCR stimulation, IL-12 signaling potentiates the stable functional IFN-γ memory in effector/memory Th1 cells characterized by hypomethylation at the IFNG promoter and CNS-1.
Collapse
Affiliation(s)
- Jun Dong
- Regenerative Immunology and Aging, Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Duverger A, Wolschendorf F, Zhang M, Wagner F, Hatcher B, Jones J, Cron RQ, van der Sluis RM, Jeeninga RE, Berkhout B, Kutsch O. An AP-1 binding site in the enhancer/core element of the HIV-1 promoter controls the ability of HIV-1 to establish latent infection. J Virol 2013; 87:2264-77. [PMID: 23236059 PMCID: PMC3571467 DOI: 10.1128/jvi.01594-12] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 12/01/2012] [Indexed: 12/13/2022] Open
Abstract
Following integration, HIV-1 in most cases produces active infection events; however, in some rare instances, latent infection events are established. The latter have major clinical implications, as latent infection allows the virus to persist despite antiretroviral therapy. Both the cellular factors and the viral elements that potentially determine whether HIV-1 establishes active or latent infection events remain largely elusive. We detail here the contribution of different long terminal repeat (LTR) sequences for the establishment of latent HIV-1 infection. Using a panel of full-length replication-competent virus constructs that reflect naturally occurring differences of HIV-1 subtype-specific LTRs and targeted LTR mutants, we found the primary ability of HIV-1 to establish latent infection in this system to be controlled by a four-nucleotide (nt) AP-1 element just upstream of the NF-κB element in the viral promoter. Deletion of this AP-1 site mostly deprived HIV-1 of the ability to establish latent HIV-1 infection. Extension of this site to a 7-nt AP-1 sequence massively promoted latency establishment, suggesting that this promoter region represents a latency establishment element (LEE). Given that these minimal changes in a transcription factor binding site affect latency establishment to such large extent, our data support the notion that HIV-1 latency is a transcription factor restriction phenomenon.
Collapse
Affiliation(s)
| | | | | | | | - Brandon Hatcher
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | - Renee M. van der Sluis
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Rienk E. Jeeninga
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Ben Berkhout
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
35
|
|
36
|
Wang C, Sandling JK, Hagberg N, Berggren O, Sigurdsson S, Karlberg O, Rönnblom L, Eloranta ML, Syvänen AC. Genome-wide profiling of target genes for the systemic lupus erythematosus-associated transcription factors IRF5 and STAT4. Ann Rheum Dis 2013; 72:96-103. [PMID: 22730365 DOI: 10.1136/annrheumdis-2012-201364] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND The transcription factors interferon regulatory factor 5 (IRF5) and signal transducer and activator of transcription 4 (STAT4) are encoded by two of the strongest susceptibility genes for systemic lupus erythematosus (SLE). OBJECTIVE To investigate the target genes and functional roles of IRF5 and STAT4 in human peripheral blood mononuclear cells (PBMCs). METHODS Chromatin immunoprecipitation-sequencing (ChIP-seq) was performed in PBMCs stimulated to activate IRF5 and STAT4. The expression of the target genes of IRF5 and STAT4 was investigated in a publicly available dataset generated from PBMCs from patients with SLE and healthy controls. The genomic regions bound by the transcription complexes mediated by IRF5 and STAT4 were examined for transcription factor binding motifs and SLE-associated sequence variants. RESULTS More than 7000 target genes for IRF5 and STAT4 were identified in stimulated PBMCs. These genes were enriched to functional pathways in the type I interferon system, and have key roles in the inflammatory response. The expression patterns of the target genes were characteristic for patients with SLE. The transcription factors high mobility group-I/Y, specificity protein 1, and paired box 4 may function cooperatively with IRF5 and STAT4 in transcriptional regulation. Eight of the target regions for IRF5 and STAT4 contain SLE-associated sequence variants. CONCLUSIONS By participating in transcription complex with other co-factors, IRF5 and STAT4 harbour the potential of regulating a large number of target genes, which may contribute to their strong association with SLE.
Collapse
Affiliation(s)
- Chuan Wang
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Tahvanainen J, Kyläniemi MK, Kanduri K, Gupta B, Lähteenmäki H, Kallonen T, Rajavuori A, Rasool O, Koskinen PJ, Rao KVS, Lähdesmäki H, Lahesmaa R. Proviral integration site for Moloney murine leukemia virus (PIM) kinases promote human T helper 1 cell differentiation. J Biol Chem 2012; 288:3048-58. [PMID: 23209281 PMCID: PMC3561529 DOI: 10.1074/jbc.m112.361709] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The differentiation of human primary T helper 1 (Th1) cells from naïve precursor cells is regulated by a complex, interrelated signaling network. The identification of factors regulating the early steps of Th1 cell polarization can provide important insight in the development of therapeutics for many inflammatory and autoimmune diseases. The serine/threonine-specific proviral integration site for Moloney murine leukemia virus (PIM) kinases PIM1 and PIM2 have been implicated in the cytokine-dependent proliferation and survival of lymphocytes. We have established that the third member of this family, PIM3, is also expressed in human primary Th cells and identified a new function for the entire PIM kinase family in T lymphocytes. Although PIM kinases are expressed more in Th1 than Th2 cells, we demonstrate here that these kinases positively influence Th1 cell differentiation. Our RNA interference results from human primary Th cells also suggest that PIM kinases promote the production of IFNγ, the hallmark cytokine produced by Th1 cells. Consistent with this, they also seem to be important for the up-regulation of the critical Th1-driving factor, T box expressed in T cells (T-BET), and the IL-12/STAT4 signaling pathway during the early Th1 differentiation process. In summary, we have identified PIM kinases as new regulators of human primary Th1 cell differentiation, thus providing new insights into the mechanisms controlling the selective development of human Th cell subsets.
Collapse
Affiliation(s)
- Johanna Tahvanainen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, 20520 Turku, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Koch M, Thomas K, Perdue N, Smigiel K, Srivastava S, Campbell D. T-bet(+) Treg cells undergo abortive Th1 cell differentiation due to impaired expression of IL-12 receptor β2. Immunity 2012; 37:501-10. [PMID: 22960221 PMCID: PMC3501343 DOI: 10.1016/j.immuni.2012.05.031] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 04/10/2012] [Accepted: 05/29/2012] [Indexed: 12/13/2022]
Abstract
Foxp3+ regulatory T (Treg) cells limit inflammatory responses and maintain immune homeostasis. Although comprised of several phenotypically and functionally distinct subsets, the differentiation of specialized Treg cell populations within the periphery is poorly characterized. We demonstrate that the development of T-bet+ Treg cells that potently inhibit T helper 1 (Th1) cell responses was dependent on the transcription factor STAT1 and occurred directly in response to interferon-γ produced by effector T cells. Additionally, delayed induction of the IL-12Rβ2 receptor component after STAT1 activation helped ensure that Treg cells do not readily complete STAT4-dependent Th1 cell development and lose their ability to suppress effector T cell proliferation. Thus, we define a pathway of abortive Th1 cell development that results in the specialization of peripheral Treg cells and demonstrate that impaired expression of a single cytokine receptor helps maintain Treg cell-suppressive function in the context of inflammatory Th1 cell responses.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Proliferation/drug effects
- Cells, Cultured
- Flow Cytometry
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interferon-gamma/pharmacology
- Interleukin-12/pharmacology
- Interleukin-12 Receptor beta 2 Subunit/genetics
- Interleukin-12 Receptor beta 2 Subunit/immunology
- Interleukin-12 Receptor beta 2 Subunit/metabolism
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, CXCR3/genetics
- Receptors, CXCR3/immunology
- Receptors, CXCR3/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- STAT1 Transcription Factor/genetics
- STAT1 Transcription Factor/immunology
- STAT1 Transcription Factor/metabolism
- STAT4 Transcription Factor/immunology
- STAT4 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Signal Transduction/immunology
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/immunology
- T-Box Domain Proteins/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Time Factors
Collapse
Affiliation(s)
- Meghan A. Koch
- Benaroya Research Institute, Seattle, WA 98101, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | | - Kate S. Smigiel
- Benaroya Research Institute, Seattle, WA 98101, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Shivani Srivastava
- Benaroya Research Institute, Seattle, WA 98101, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Daniel J. Campbell
- Benaroya Research Institute, Seattle, WA 98101, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Corresponding author
| |
Collapse
|
39
|
Dykhuizen EC, Carmody LC, Tolliday N, Crabtree GR, Palmer MAJ. Screening for inhibitors of an essential chromatin remodeler in mouse embryonic stem cells by monitoring transcriptional regulation. ACTA ACUST UNITED AC 2012; 17:1221-30. [PMID: 22853929 DOI: 10.1177/1087057112455060] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The SWI/SNF-like adenosine triphosphate (ATP)-dependent chromatin remodeling complex, esBAF, is both necessary and, in some contexts, sufficient to induce the pluripotent state. Furthermore, mutations in various BAF subunits are associated with cancer. Little is known regarding the precise mechanism(s) by which this complex exerts its activities. Thus, it is unclear which protein interactions would be important to disrupt to isolate a relevant readout of mechanism. To address this, we developed a gene expression-based assay to identify inhibitors of the native esBAF complex. Specifically, a quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) assay was developed in mouse embryonic stem (ES) cells to monitor expression of Bmi1, a developmentally important gene repressed by the esBAF complex. The assay was miniaturized to a 384-well format and used to screen a diverse collection of compounds, including novel products of diversity-oriented synthesis (DOS). Confirmed hits were validated using a knock-in ES cell reporter line in which luciferase is inserted into the Bmi1 locus. Several of the validated hits regulate a panel of target genes in a manner similar to the BAF chromatin-remodeling complex. Together these data indicate that expression-based screening using qRT-PCR is a successful approach to identify compounds targeting the regulation of key developmental genes in ES cells.
Collapse
Affiliation(s)
- Emily C Dykhuizen
- Department of Pathology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | | | | | |
Collapse
|
40
|
Verma VK, Taneja V, Jaiswal A, Sharma S, Behera D, Sreenivas V, Chauhan SS, Prasad HK. Prevalence, distribution and functional significance of the -237C to T polymorphism in the IL-12Rβ2 promoter in Indian tuberculosis patients. PLoS One 2012; 7:e34355. [PMID: 22509293 PMCID: PMC3317943 DOI: 10.1371/journal.pone.0034355] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 02/27/2012] [Indexed: 01/03/2023] Open
Abstract
Cytokine/cytokine receptor gene polymorphisms related to structure/expression could impact immune response. Hence, the −237 polymorphic site in the 5′ promoter region of the IL-12Rβ2 (SNP ID: rs11810249) gene associated with the AP-4 transcription motif GAGCTG, was examined. Amplicons encompassing the polymorphism were generated from 46 pulmonary tuberculosis patients, 35 family contacts and 28 miscellaneous volunteers and sequenced. The C allele predominated among patients, (93.4%, 43/46), and in all volunteers and contacts screened, but the T allele was exclusively limited to patients, (6.5%, 3/46). The functional impact of this polymorphism on transcriptional activity was assessed by Luciferase-reporter and electrophoretic mobility shift assays (EMSA). Luciferase-reporter assays showed a significant reduction in transcriptional efficiency with T compared to C allele. The reduction in transcriptional efficiency with the T allele construct (pGIL-12Rb2-T), in U-87MG, THP-1 and Jurkat cell lines, were 53, 37.6, and 49.8% respectively, compared to the C allele construct (pGIL-12Rb2-C). Similarly, densitometric analysis of the EMSA assay showed reduced binding of the AP-4 transcription factor, to T compared to the C nucleotide probe. Reduced mRNA expression in all patients (3/3) harboring the T allele was seen, whereas individuals with the C allele exhibited high mRNA expression (17/25; 68%, p = 0.05). These observations were in agreement with the in vitro assessment of the promoter activity by Luciferase-reporter and EMSA assays. The reduced expression of IL-12Rβ2 transcripts in 8 patients despite having the C allele was attributed to the predominant over expression of the suppressors (IL-4 and GATA-3) and reduced expression of enhancers (IFN-α) of IL-12Rβ2 transcripts. The 17 high IL-12Rβ2 mRNA expressers had significantly elevated IFN-α mRNA levels compared to low expressers and volunteers. Notwithstanding the presence of high levels of IL-12Rβ2 mRNA in these patients elevated IFN-α expression could modulate their immune responses to Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Vikas Kumar Verma
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Vibha Taneja
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Anand Jaiswal
- LRS Institute of Tuberculosis and Respiratory Diseases, New Delhi, India
| | - Sangeeta Sharma
- LRS Institute of Tuberculosis and Respiratory Diseases, New Delhi, India
| | - Digamber Behera
- LRS Institute of Tuberculosis and Respiratory Diseases, New Delhi, India
| | - Vishnubhatla Sreenivas
- Department of Biostatistics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Shyam Singh Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | |
Collapse
|
41
|
Abstract
One of the best studied systems for mammalian chromatin remodeling is transcriptional regulation during T cell development. The variety of these studies have led to important findings in T cell gene regulation and cell fate determination. Importantly, these findings have also advanced our knowledge of the function of remodeling enzymes in mammalian gene regulation. First we briefly present biochemical and cell-free analysis of 3 types of ATP dependent remodeling enzymes (SWI/SNF, Mi2, and ISWI) to construct an intellectual framework to understand how these enzymes might be working. Second, we compare and contrast the function of these enzymes during early (thymic) and late (peripheral) T cell development. Finally, we examine some of the gaps in our present understanding.
Collapse
Affiliation(s)
- Andrea L. Wurster
- Laboratory of Molecular Biology and Immunology, National Institute on Aging Intramural Research Program, National Institutes of Health, USA
| | - Michael J. Pazin
- Laboratory of Molecular Biology and Immunology, National Institute on Aging Intramural Research Program, National Institutes of Health, USA
| |
Collapse
|
42
|
Wurster AL, Precht P, Pazin MJ. NF-κB and BRG1 bind a distal regulatory element in the IL-3/GM-CSF locus. Mol Immunol 2011; 48:2178-88. [PMID: 21831442 DOI: 10.1016/j.molimm.2011.07.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 07/17/2011] [Accepted: 07/19/2011] [Indexed: 01/15/2023]
Abstract
We investigated gene regulation at the IL-3/GM-CSF gene cluster. We found BRG1, a SWI/SNF remodeling ATPase, bound a distal element, CNSa. BRG1 binding was strongest in differentiated, stimulated T helper cells, paralleling IL-3 and GM-CSF expression. Depletion of BRG1 reduced IL-3 and GM-CSF transcription. BAF-specific SWI/SNF subunits bound to this locus and regulated IL-3 expression. CNSa was in closed chromatin in fibroblasts, open chromatin in differentiated T helper cells, and moderately open chromatin in naïve (undifferentiated) T helper cells; BRG1 was required for the most open state. CNSa increased transcription of a reporter in an episomal expression system, in a BRG1-dependent manner. The NF-κB subunit RelA/p65 bound CNSa in activated T helper cells. Inhibition of NF-κB blocked BRG1 binding to CNSa, chromatin opening at CNSa, and activation of IL-3 and GM-CSF. Together, these findings suggest CNSa is a distal enhancer that binds BRG1 and NF-κB.
Collapse
Affiliation(s)
- Andrea L Wurster
- Laboratory of Molecular Biology and Immunology, National Institute on Aging Intramural Research Program, National Institutes of Health, USA
| | | | | |
Collapse
|
43
|
De S, Wurster AL, Precht P, Wood WH, Becker KG, Pazin MJ. Dynamic BRG1 recruitment during T helper differentiation and activation reveals distal regulatory elements. Mol Cell Biol 2011; 31:1512-27. [PMID: 21262765 PMCID: PMC3135292 DOI: 10.1128/mcb.00920-10] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 01/14/2011] [Indexed: 12/12/2022] Open
Abstract
T helper cell differentiation and activation require specific transcriptional programs accompanied by changes in chromatin structure. However, little is known about the chromatin remodeling enzymes responsible. We performed genome-wide analysis to determine the general principles of BRG1 binding, followed by analysis of specific genes to determine whether these general rules were typical of key T cell genes. We found that binding of the remodeling protein BRG1 was programmed by both lineage and activation signals. BRG1 binding positively correlated with gene activity at protein-coding and microRNA (miRNA) genes. BRG1 binding was found at promoters and distal regions, including both novel and previously validated distal regulatory elements. Distal BRG1 binding correlated with expression, and novel distal sites in the Gata3 locus possessed enhancer-like activity, suggesting a general role for BRG1 in long-distance gene regulation. BRG1 recruitment to distal sites in Gata3 was impaired in cells lacking STAT6, a transcription factor that regulates lineage-specific genes. Together, these findings suggest that BRG1 interprets both differentiation and activation signals and plays a causal role in gene regulation, chromatin structure, and cell fate. Our findings suggest that BRG1 binding is a useful marker for identifying active cis-regulatory regions in protein-coding and miRNA genes.
Collapse
Affiliation(s)
| | - Andrea L. Wurster
- Laboratory of Cellular and Molecular Biology, National Institute on Aging, NIH, Baltimore, Maryland 21224
| | - Patricia Precht
- Laboratory of Cellular and Molecular Biology, National Institute on Aging, NIH, Baltimore, Maryland 21224
| | | | | | - Michael J. Pazin
- Laboratory of Cellular and Molecular Biology, National Institute on Aging, NIH, Baltimore, Maryland 21224
| |
Collapse
|
44
|
Precht P, Wurster AL, Pazin MJ. The SNF2H chromatin remodeling enzyme has opposing effects on cytokine gene expression. Mol Immunol 2010; 47:2038-46. [PMID: 20471682 PMCID: PMC2891439 DOI: 10.1016/j.molimm.2010.04.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 04/13/2010] [Accepted: 04/14/2010] [Indexed: 01/10/2023]
Abstract
Cytokine gene expression is a key control point in the function of the immune system. Cytokine gene regulation is linked to changes in chromatin structure; however, little is known about the remodeling enzymes mediating these changes. Here we investigated the role of the ATP-dependent chromatin remodeling enzyme SNF2H in mouse T cells; to date, SNF2H has not been investigated in T cells. We found that SNF2H repressed expression of IL-2 and other cytokines in activated cells. By contrast, SNF2H activated expression of IL-3. The ISWI components SNF2H and ACF1 bound to the tested loci, suggesting the regulation was direct. SNF2H decreased accessibility at some binding sites within the IL2 locus, and increased accessibility within some IL3 binding sites. The changes in gene expression positively correlated with accessibility changes, suggesting a simple model that accessibility enables transcription. We also found that loss of the ISWI ATPase SNF2H reduced binding to target genes and protein expression of ACF1, a binding partner for SNF2H, suggesting complex formation stabilized ACF1. Together, these findings reveal a direct role for SNF2H in both repression and activation of cytokine genes.
Collapse
Affiliation(s)
- Patricia Precht
- Laboratory of Cellular and Molecular Biology National Institute on Aging, NIH Baltimore, MD 21224
| | - Andrea L. Wurster
- Laboratory of Cellular and Molecular Biology National Institute on Aging, NIH Baltimore, MD 21224
| | - Michael J. Pazin
- Address correspondence and reprint requests to: Dr. Michael J. Pazin; LCMB. NIA, NIH; 251 Bayview Blvd; Baltimore, MD 21224. Phone: 410-558-8094; Fax: 410-558-8386;
| |
Collapse
|
45
|
Abstract
Immunological memory is a cardinal feature of adaptive immunity. We are now beginning to elucidate the mechanisms that govern the formation of memory T cells and their ability to acquire longevity, survive the effector-to-memory transition, and mature into multipotent, functional memory T cells that self-renew. Here, we discuss the recent findings in this area and highlight extrinsic and intrinsic factors that regulate the cellular fate of activated CD8+ T cells.
Collapse
Affiliation(s)
- Weiguo Cui
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Susan M. Kaech
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
46
|
Wei L, Vahedi G, Sun HW, Watford WT, Takatori H, Ramos HL, Takahashi H, Liang J, Gutierrez-Cruz G, Zang C, Peng W, O'Shea JJ, Kanno Y. Discrete roles of STAT4 and STAT6 transcription factors in tuning epigenetic modifications and transcription during T helper cell differentiation. Immunity 2010; 32:840-51. [PMID: 20620946 PMCID: PMC2904651 DOI: 10.1016/j.immuni.2010.06.003] [Citation(s) in RCA: 257] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 04/20/2010] [Accepted: 05/06/2010] [Indexed: 01/30/2023]
Abstract
Signal transducer and activator of transcription 4 (STAT4) and STAT6 are key factors in the specification of helper T cells; however, their direct roles in driving differentiation are not well understood. Using chromatin immunoprecipitation and massive parallel sequencing, we quantitated the full complement of STAT-bound genes, concurrently assessing global STAT-dependent epigenetic modifications and gene transcription by using cells from cognate STAT-deficient mice. STAT4 and STAT6 each bound over 4000 genes with distinct binding motifs. Both played critical roles in maintaining chromatin configuration and transcription of a core subset of genes through the combination of different epigenetic patterns. Globally, STAT4 had a more dominant role in promoting active epigenetic marks, whereas STAT6 had a more prominent role in antagonizing repressive marks. Clusters of genes negatively regulated by STATs were also identified, highlighting previously unappreciated repressive roles of STATs. Therefore, STAT4 and STAT6 play wide regulatory roles in T helper cell specification.
Collapse
Affiliation(s)
- Lai Wei
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Golnaz Vahedi
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Wendy T. Watford
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Hiroaki Takatori
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Haydee L. Ramos
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Hayato Takahashi
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Jonathan Liang
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Gustavo Gutierrez-Cruz
- Laboratory of Muscle Stem Cells and Gene Regulation, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Chongzhi Zang
- Department of Physics, George Washington University, Washington, DC 20052, U.S.A
| | - Weiqun Peng
- Department of Physics, George Washington University, Washington, DC 20052, U.S.A
| | - John J. O'Shea
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| | - Yuka Kanno
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, U.S.A
| |
Collapse
|
47
|
In vivo expansion of naive and activated CD4+CD25+FOXP3+ regulatory T cell populations in interleukin-2-treated HIV patients. Proc Natl Acad Sci U S A 2010; 107:10632-7. [PMID: 20498045 DOI: 10.1073/pnas.1000027107] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
HIV-1 infection is characterized by a progressive decline in CD4(+) T cells leading to a state of profound immunodeficiency. IL-2 therapy has been shown to improve CD4(+) counts beyond that observed with antiretroviral therapy. Recent phase III trials revealed that despite a sustained increase in CD4(+) counts, IL-2-treated patients did not experience a better clinical outcome [Abrams D, et al. (2009) N Engl J Med 361(16):1548-1559]. To explain these disappointing results, we have studied phenotypic, functional, and molecular characteristics of CD4(+) T cell populations in IL-2-treated patients. We found that the principal effect of long-term IL-2 therapy was the expansion of two distinct CD4(+)CD25(+) T cell populations (CD4(+)CD25(lo)CD127(lo)FOXP3(+) and CD4(+)CD25(hi)CD127(lo)FOXP3(hi)) that shared phenotypic markers of Treg but could be distinguished by the levels of CD25 and FOXP3 expression. IL-2-expanded CD4(+)CD25(+) T cells suppressed proliferation of effector cells in vitro and had gene expression profiles similar to those of natural regulatory CD4(+)CD25(hi)FOXP3(+) T cells (Treg) from healthy donors, an immunosuppressive T cell subset critically important for the maintenance of self-tolerance. We propose that the sustained increase of the peripheral Treg pool in IL-2-treated HIV patients may account for the unexpected clinical observation that patients with the greatest expansion of CD4(+) T cells had a higher relative risk of clinical progression to AIDS.
Collapse
|
48
|
Placek K, Gasparian S, Coffre M, Maiella S, Sechet E, Bianchi E, Rogge L. Integration of distinct intracellular signaling pathways at distal regulatory elements directs T-bet expression in human CD4+ T cells. THE JOURNAL OF IMMUNOLOGY 2010; 183:7743-51. [PMID: 19923468 DOI: 10.4049/jimmunol.0803812] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
T-bet is a key regulator controlling Th1 cell development. This factor is not expressed in naive CD4(+) T cells, and the mechanisms controlling expression of T-bet are incompletely understood. In this study, we defined regulatory elements at the human T-bet locus and determined how signals originating at the TCR and at cytokine receptors are integrated to induce chromatin modifications and expression of this gene during human Th1 cell differentiation. We found that T cell activation induced two strong DNase I-hypersensitive sites (HS) and rapid histone acetylation at these elements in CD4(+) T cells. Histone acetylation and T-bet expression were strongly inhibited by cyclosporine A, and we detected binding of NF-AT to a HS in vivo. IL-12 and IFN-gamma signaling alone were not sufficient to induce T-bet expression in naive CD4(+) T cells, but enhanced T-bet expression in TCR/CD28-stimulated cells. We detected a third HS 12 kb upstream of the mRNA start site only in developing Th1 cells, which was bound by IL-12-induced STAT4. Our data suggest that T-bet locus remodeling and gene expression are initiated by TCR-induced NF-AT recruitment and amplified by IL-12-mediated STAT4 binding to distinct distal regulatory elements during human Th1 cell differentiation.
Collapse
Affiliation(s)
- Katarzyna Placek
- Institut Pasteur, Immunoregulation Unit and Centre National de la Recherche Scientifique Unité de Recherche Associée 1961, Department of Immunology, Paris, France
| | | | | | | | | | | | | |
Collapse
|
49
|
NOMA T. Helper T cell paradigm : Th17 and regulatory T cells involved in autoimmune inflammatory disorders, pathogen defense and allergic diseases. ACTA ACUST UNITED AC 2010; 33:262-71. [DOI: 10.2177/jsci.33.262] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Takeshi NOMA
- Kitasato University Graduate School of Medical Science, Department of Pediatrics
| |
Collapse
|
50
|
Jeong SM, Lee C, Lee SK, Kim J, Seong RH. The SWI/SNF chromatin-remodeling complex modulates peripheral T cell activation and proliferation by controlling AP-1 expression. J Biol Chem 2009; 285:2340-50. [PMID: 19910461 DOI: 10.1074/jbc.m109.026997] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The SWI/SNF chromatin-remodeling complex has been implicated in the activation and proliferation of T cells. After T cell receptor signaling, the SWI/SNF complex rapidly associates with chromatin and controls gene expression in T cells. However, the process by which the SWI/SNF complex regulates peripheral T cell activation has not been elucidated. In this study, we show that the SWI/SNF complex regulates cytokine production and proliferation of T cells. During T cell activation, the SWI/SNF complex is recruited to the promoter of the transcription factor AP-1, and it increases the expression of AP-1. Increased expression of the SWI/SNF complex resulted in enhanced AP-1 activity, cytokine production, and proliferation of peripheral T cells, whereas knockdown of the SWI/SNF complex expression impaired the AP-1 expression and reduced the activation and proliferation of T cells. Moreover, mice that constitutively expressed the SWI/SNF complex in T cells were much more susceptible to experimentally induced autoimmune encephalomyelitis than the normal mice were. These results suggest that the SWI/SNF complex plays a critical role during T cell activation and subsequent immune responses.
Collapse
Affiliation(s)
- Seung Min Jeong
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, and Research Center for Functional Cellulomics, Seoul National University, 599 Gwanangno, Gwanak-gu, Seoul 151-742, Korea
| | | | | | | | | |
Collapse
|