1
|
Sobhy H, De Rovere M, Ait-Ammar A, Kashif M, Wallet C, Daouad F, Loustau T, Van Lint C, Schwartz C, Rohr O. BCL11b interacts with RNA and proteins involved in RNA processing and developmental diseases. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195065. [PMID: 39455000 DOI: 10.1016/j.bbagrm.2024.195065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
BCL11b is a transcription regulator and a tumor suppressor involved in lymphomagenesis, central nervous system (CNS) and immune system developments. BCL11b favors persistence of HIV latency and contributes to control cell cycle, differentiation and apoptosis in multiple organisms and cell models. Although BCL11b recruits the non-coding RNA 7SK and epigenetic enzymes to regulate gene expression, BCL11b-associated ribonucleoprotein complexes are unknown. Thanks to CLIP-seq and quantitative LC-MS/MS mass spectrometry approaches complemented with systems biology validations, we show that BCL11b interacts with RNA splicing and non-sense-mediated decay proteins, including FUS, SMN1, UPF1 and Drosha, which may contribute in isoform selection of protein-coding RNA isoforms from noncoding-RNAs isoforms (retained introns or nonsense mediated RNA). Interestingly, BCL11b binds to RNA transcripts and proteins encoded by the same genes (FUS, ESWR1, CHD and Tubulin). Our study highlights that BCL11b targets RNA processing and splicing proteins, and RNAs that implicate cell cycle, development, neurodegenerative, and cancer pathways. These findings will help future mechanistic understanding of developmental disorders. IMPORTANCE: BCL11b-protein and RNA interactomes reveal BLC11b association with specific nucleoprotein complexes involved in the regulation of genes expression. BCL11b interacts with RNA processing and splicing proteins.
Collapse
Affiliation(s)
- Haitham Sobhy
- University of Strasbourg, UR 7292, DHPI, IUT Louis Pasteur, Schiltigheim, France.
| | - Marco De Rovere
- University of Strasbourg, UR 7292, DHPI, IUT Louis Pasteur, Schiltigheim, France
| | - Amina Ait-Ammar
- University of Strasbourg, UR 7292, DHPI, IUT Louis Pasteur, Schiltigheim, France; Université Libre de Bruxelles, ULB, Gosselies, Belgium
| | - Muhammad Kashif
- University of Strasbourg, UPR CNRS 9002, ARN, IUT Louis Pasteur, Schiltigheim, France
| | - Clementine Wallet
- University of Strasbourg, UPR CNRS 9002, ARN, IUT Louis Pasteur, Schiltigheim, France
| | - Fadoua Daouad
- University of Strasbourg, UR 7292, DHPI, IUT Louis Pasteur, Schiltigheim, France
| | - Thomas Loustau
- University of Strasbourg, UPR CNRS 9002, ARN, IUT Louis Pasteur, Schiltigheim, France
| | | | - Christian Schwartz
- University of Strasbourg, UPR CNRS 9002, ARN, IUT Louis Pasteur, Schiltigheim, France
| | - Olivier Rohr
- University of Strasbourg, UPR CNRS 9002, ARN, IUT Louis Pasteur, Schiltigheim, France.
| |
Collapse
|
2
|
Meng L, Huo Z. Outcome-guided Bayesian clustering for disease subtype discovery using high-dimensional transcriptomic data. J Appl Stat 2024; 52:183-207. [PMID: 39811087 PMCID: PMC11727188 DOI: 10.1080/02664763.2024.2362275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/23/2024] [Indexed: 01/16/2025]
Abstract
Due to the tremendous heterogeneity of disease manifestations, many complex diseases that were once thought to be single diseases are now considered to have disease subtypes. Disease subtyping analysis, that is the identification of subgroups of patients with similar characteristics, is the first step to accomplish precision medicine. With the advancement of high-throughput technologies, omics data offers unprecedented opportunity to reveal disease subtypes. As a result, unsupervised clustering analysis has been widely used for this purpose. Though promising, the subtypes obtained from traditional quantitative approaches may not always be clinically meaningful (i.e. correlate with clinical outcomes). On the other hand, the collection of rich clinical data in modern epidemiology studies has the great potential to facilitate the disease subtyping process via omics data and to discovery clinically meaningful disease subtypes. Thus, we developed an outcome-guided Bayesian clustering (GuidedBayesianClustering) method to fully integrate the clinical data and the high-dimensional omics data. A Gaussian mixed model framework was applied to perform sample clustering; a spike-and-slab prior was utilized to perform gene selection; a mixture model prior was employed to incorporate the guidance from a clinical outcome variable; and a decision framework was adopted to infer the false discovery rate of the selected genes. We deployed conjugate priors to facilitate efficient Gibbs sampling. Our proposed full Bayesian method is capable of simultaneously (i) obtaining sample clustering (disease subtype discovery); (ii) performing feature selection (select genes related to the disease subtype); and (iii) utilizing clinical outcome variable to guide the disease subtype discovery. The superior performance of the GuidedBayesianClustering was demonstrated through simulations and applications of breast cancer expression data and Alzheimer's disease. An R package has been made publicly available on GitHub to improve the applicability of our method.
Collapse
Affiliation(s)
- Lingsong Meng
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
3
|
Sha J, Zhang M, Feng J, Shi T, Li N, Jie Z. Promyelocytic leukemia zinc finger controls type 2 immune responses in the lungs by regulating lineage commitment and the function of innate and adaptive immune cells. Int Immunopharmacol 2024; 130:111670. [PMID: 38373386 DOI: 10.1016/j.intimp.2024.111670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
Type 2 immune responses are critical for host defense, mediate allergy and Th2-high asthma. The transcription factor, promyelocytic leukemia zinc finger (PLZF), has emerged as a significant regulator of type 2 inflammation in the lung; however, its exact mechanism remains unclear. In this review, we summarized recent findings regarding the ability of PLZF to control the development and function of innate lymphoid cells (ILCs), iNKT cells, memory T cells, basophils, and other immune cells that drive type 2 responses. We discussed the important role of PLZF in the pathogenesis of Th2-high asthma. Collectively, prior studies have revealed the critical role of PLZF in the regulation of innate and adaptive immune cells involved in type 2 inflammation in the lung. Therefore, targeting PLZF signaling represents a promising therapeutic approach to suppress Th2-high asthma.
Collapse
Affiliation(s)
- Jiafeng Sha
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jingjing Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Tianyun Shi
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Na Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Zhijun Jie
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Center of Community-Based Health Research, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Gray EH, Srenathan U, Durham LE, Lalnunhlimi S, Steel KJA, Catrina A, Kirkham BW, Taams LS. Human in vitro-induced IL-17A+ CD8+ T-cells exert pro-inflammatory effects on synovial fibroblasts. Clin Exp Immunol 2023; 214:103-119. [PMID: 37367825 PMCID: PMC10711358 DOI: 10.1093/cei/uxad068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/25/2023] [Accepted: 06/26/2023] [Indexed: 06/28/2023] Open
Abstract
IL-17A+ CD8+ T-cells, termed Tc17 cells, have been identified at sites of inflammation in several immune-mediated inflammatory diseases. However, the biological function of human IL-17A+ CD8+ T-cells is not well characterized, likely due in part to the relative scarcity of these cells. Here, we expanded IL-17A+ CD8+ T-cells from healthy donor PBMC or bulk CD8+ T-cell populations using an in vitro polarization protocol. We show that T-cell activation in the presence of IL-1β and IL-23 significantly increased the frequencies of IL-17A+ CD8+ T-cells, which was not further enhanced by IL-6, IL-2, or anti-IFNγ mAb addition. In vitro-generated IL-17A+ CD8+ T-cells displayed a distinct type-17 profile compared with IL-17A- CD8+ T-cells, as defined by transcriptional signature (IL17A, IL17F, RORC, RORA, MAF, IL23R, CCR6), high surface expression of CCR6 and CD161, and polyfunctional production of IL-17A, IL-17F, IL-22, IFNγ, TNFα, and GM-CSF. A significant proportion of in vitro-induced IL-17A+ CD8+ T-cells expressed TCRVα7.2 and bound MR1 tetramers indicative of MAIT cells, indicating that our protocol expanded both conventional and unconventional IL-17A+ CD8+ T-cells. Using an IL-17A secretion assay, we sorted the in vitro-generated IL-17A+ CD8+ T-cells for functional analysis. Both conventional and unconventional IL-17A+ CD8+ T-cells were able to induce pro-inflammatory IL-6 and IL-8 production by synovial fibroblasts from patients with psoriatic arthritis, which was reduced upon addition of anti-TNFα and anti-IL-17A neutralizing antibodies. Collectively, these data demonstrate that human in vitro-generated IL-17A+ CD8+ T-cells are biologically functional and that their pro-inflammatory function can be targeted, at least in vitro, using existing immunotherapy.
Collapse
Affiliation(s)
- Elizabeth H Gray
- Centre for Inflammation Biology and Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Ushani Srenathan
- Centre for Inflammation Biology and Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Lucy E Durham
- Centre for Inflammation Biology and Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Sylvine Lalnunhlimi
- Centre for Inflammation Biology and Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Kathryn J A Steel
- Centre for Inflammation Biology and Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, London, UK
| | - Anca Catrina
- Rheumatology Unit, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Bruce W Kirkham
- Department of Rheumatology, Guy’s Hospital, Guy’s and St. Thomas’ NHS Foundation Trust Hospital, London, UK
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, London, UK
| |
Collapse
|
5
|
Chandra S, Ascui G, Riffelmacher T, Chawla A, Ramírez-Suástegui C, Castelan VC, Seumois G, Simon H, Murray MP, Seo GY, Premlal ALR, Schmiedel B, Verstichel G, Li Y, Lin CH, Greenbaum J, Lamberti J, Murthy R, Nigro J, Cheroutre H, Ottensmeier CH, Hedrick SM, Lu LF, Vijayanand P, Kronenberg M. Transcriptomes and metabolism define mouse and human MAIT cell populations. Sci Immunol 2023; 8:eabn8531. [PMID: 37948512 PMCID: PMC11160507 DOI: 10.1126/sciimmunol.abn8531] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells are a subset of T lymphocytes that respond to microbial metabolites. We defined MAIT cell populations in different organs and characterized the developmental pathway of mouse and human MAIT cells in the thymus using single-cell RNA sequencing and phenotypic and metabolic analyses. We showed that the predominant mouse subset, which produced IL-17 (MAIT17), and the subset that produced IFN-γ (MAIT1) had not only greatly different transcriptomes but also different metabolic states. MAIT17 cells in different organs exhibited increased lipid uptake, lipid storage, and mitochondrial potential compared with MAIT1 cells. All these properties were similar in the thymus and likely acquired there. Human MAIT cells in lung and blood were more homogeneous but still differed between tissues. Human MAIT cells had increased fatty acid uptake and lipid storage in blood and lung, similar to human CD8 T resident memory cells, but unlike mouse MAIT17 cells, they lacked increased mitochondrial potential. Although mouse and human MAIT cell transcriptomes showed similarities for immature cells in the thymus, they diverged more strikingly in the periphery. Analysis of pet store mice demonstrated decreased lung MAIT17 cells in these so-called "dirty" mice, indicative of an environmental influence on MAIT cell subsets and function.
Collapse
Affiliation(s)
- Shilpi Chandra
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Gabriel Ascui
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093 USA
| | - Thomas Riffelmacher
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY UK
| | - Ashu Chawla
- Bioinformatics Core Facility, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Ciro Ramírez-Suástegui
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Viankail C. Castelan
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Gregory Seumois
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Hayley Simon
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Mallory P. Murray
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Goo-Young Seo
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | | | - Benjamin Schmiedel
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Greet Verstichel
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Yingcong Li
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
| | - Chia-Hao Lin
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
| | - Jason Greenbaum
- Bioinformatics Core Facility, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - John Lamberti
- Division of Cardiac Surgery, Rady Children’s Hospital, San Diego, CA 92123 USA
- Division of Pediatric Cardiac Surgery, Falk Cardiovascular Research Center, Stanford, CA 94305-5407 USA
| | - Raghav Murthy
- Division of Cardiac Surgery, Rady Children’s Hospital, San Diego, CA 92123 USA
- Division of Pediatric Cardiac Surgery, Children’s Heart Center Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - John Nigro
- Division of Cardiac Surgery, Rady Children’s Hospital, San Diego, CA 92123 USA
| | - Hilde Cheroutre
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Christian H. Ottensmeier
- Liverpool Head and Neck Center, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK, L69 7ZB
| | - Stephen M. Hedrick
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093 USA
| | - Li-Fan Lu
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, 92093 USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093 USA
| | - Pandurangan Vijayanand
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Mitchell Kronenberg
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
| |
Collapse
|
6
|
Perriman L, Tavakolinia N, Jalali S, Li S, Hickey PF, Amann-Zalcenstein D, Ho WWH, Baldwin TM, Piers AT, Konstantinov IE, Anderson J, Stanley EG, Licciardi PV, Kannourakis G, Naik SH, Koay HF, Mackay LK, Berzins SP, Pellicci DG. A three-stage developmental pathway for human Vγ9Vδ2 T cells within the postnatal thymus. Sci Immunol 2023; 8:eabo4365. [PMID: 37450574 DOI: 10.1126/sciimmunol.abo4365] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
Vγ9Vδ2 T cells are the largest population of γδ T cells in adults and can play important roles in providing effective immunity against cancer and infection. Many studies have suggested that peripheral Vγ9Vδ2 T cells are derived from the fetal liver and thymus and that the postnatal thymus plays little role in the development of these cells. More recent evidence suggested that these cells may also develop postnatally in the thymus. Here, we used high-dimensional flow cytometry, transcriptomic analysis, functional assays, and precursor-product experiments to define the development pathway of Vγ9Vδ2 T cells in the postnatal thymus. We identify three distinct stages of development for Vγ9Vδ2 T cells in the postnatal thymus that are defined by the progressive acquisition of functional potential and major changes in the expression of transcription factors, chemokines, and other surface markers. Furthermore, our analysis of donor-matched thymus and blood revealed that the molecular requirements for the development of functional Vγ9Vδ2 T cells are delivered predominantly by the postnatal thymus and not in the periphery. Tbet and Eomes, which are required for IFN-γ and TNFα expression, are up-regulated as Vγ9Vδ2 T cells mature in the thymus, and mature thymic Vγ9Vδ2 T cells rapidly express high levels of these cytokines after stimulation. Similarly, the postnatal thymus programs Vγ9Vδ2 T cells to express the cytolytic molecules, perforin, granzyme A, and granzyme K. This study provides a greater understanding of how Vγ9Vδ2 T cells develop in humans and may lead to opportunities to manipulate these cells to treat human diseases.
Collapse
Affiliation(s)
- Louis Perriman
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Fiona Elsey Cancer Research Institute, Ballarat, Australia
- Federation University Australia, Ballarat, Australia
| | - Naeimeh Tavakolinia
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Sedigheh Jalali
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Shuo Li
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Peter F Hickey
- Advanced Genomics Facility and Single Cell Open Research Endeavour (SCORE), Advanced Technology and Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Daniela Amann-Zalcenstein
- Advanced Genomics Facility and Single Cell Open Research Endeavour (SCORE), Advanced Technology and Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - William Wing Ho Ho
- Advanced Genomics Facility and Single Cell Open Research Endeavour (SCORE), Advanced Technology and Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Tracey M Baldwin
- Advanced Genomics Facility and Single Cell Open Research Endeavour (SCORE), Advanced Technology and Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Adam T Piers
- Murdoch Children's Research Institute, Melbourne, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Melbourne, Australia
| | - Igor E Konstantinov
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Melbourne, Australia
- Cardiothoracic Surgery, Royal Children's Hospital, Melbourne, Australia
| | - Jeremy Anderson
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Edouard G Stanley
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, Australia
- Federation University Australia, Ballarat, Australia
| | - Shalin H Naik
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Stuart P Berzins
- Fiona Elsey Cancer Research Institute, Ballarat, Australia
- Federation University Australia, Ballarat, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Daniel G Pellicci
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Melbourne, Australia
| |
Collapse
|
7
|
Helm EY, Zelenka T, Cismasiu VB, Islam S, Silvane L, Zitti B, Holmes TD, Drashansky TT, Kwiatkowski AJ, Tao C, Dean J, Obermayer AN, Chen X, Keselowsky BG, Zhang W, Huo Z, Zhou L, Sheridan BS, Conejo-Garcia JR, Shaw TI, Bryceson YT, Avram D. Bcl11b sustains multipotency and restricts effector programs of intestinal-resident memory CD8 + T cells. Sci Immunol 2023; 8:eabn0484. [PMID: 37115913 DOI: 10.1126/sciimmunol.abn0484] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
The networks of transcription factors (TFs) that control intestinal-resident memory CD8+ T (TRM) cells, including multipotency and effector programs, are poorly understood. In this work, we investigated the role of the TF Bcl11b in TRM cells during infection with Listeria monocytogenes using mice with post-activation, conditional deletion of Bcl11b in CD8+ T cells. Conditional deletion of Bcl11b resulted in increased numbers of intestinal TRM cells and their precursors as well as decreased splenic effector and circulating memory cells and precursors. Loss of circulating memory cells was in part due to increased intestinal homing of Bcl11b-/- circulating precursors, with no major alterations in their programs. Bcl11b-/- TRM cells had altered transcriptional programs, with diminished expression of multipotent/multifunctional (MP/MF) program genes, including Tcf7, and up-regulation of the effector program genes, including Prdm1. Bcl11b also limits the expression of Ahr, another TF with a role in intestinal CD8+ TRM cell differentiation. Deregulation of TRM programs translated into a poor recall response despite TRM cell accumulation in the intestine. Reduced expression of MP/MF program genes in Bcl11b-/- TRM cells was linked to decreased chromatin accessibility and a reduction in activating histone marks at these loci. In contrast, the effector program genes displayed increased activating epigenetic status. These findings demonstrate that Bcl11b is a frontrunner in the tissue residency program of intestinal memory cells upstream of Tcf1 and Blimp1, promoting multipotency and restricting the effector program.
Collapse
Affiliation(s)
- Eric Y Helm
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tomas Zelenka
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Valeriu B Cismasiu
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Shamima Islam
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Leonardo Silvane
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Beatrice Zitti
- Centre for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, S-14186 Stockholm, Sweden
| | - Tim D Holmes
- Broegelmann Research Laboratory, Department of Clinical Sciences, University of Bergen, N-5021 Bergen, Norway
| | - Theodore T Drashansky
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Alexander J Kwiatkowski
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Christine Tao
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Joseph Dean
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Alyssa N Obermayer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Xianghong Chen
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- UF Health Cancer Center, Gainesville, FL 32610, USA
| | - Zhiguang Huo
- Department of Biostatistics, College of Medicine, College of Public Health & Health Professions, University of Florida, Gainesville, FL 32611, USA
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Brian S Sheridan
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Timothy I Shaw
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Yenan T Bryceson
- Centre for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, S-14186 Stockholm, Sweden
- Broegelmann Research Laboratory, Department of Clinical Sciences, University of Bergen, N-5021 Bergen, Norway
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, S-14186 Stockholm, Sweden
| | - Dorina Avram
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL 33612, USA
| |
Collapse
|
8
|
Wei ZY, Wang ZX, Li JH, Wen YS, Gao D, Xia SY, Li YN, Pan XB, Liu YS, Jin YY, Chen JH. Host A-to-I RNA editing signatures in intracellular bacterial and single-strand RNA viral infections. Front Immunol 2023; 14:1121096. [PMID: 37081881 PMCID: PMC10112020 DOI: 10.3389/fimmu.2023.1121096] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/13/2023] [Indexed: 04/07/2023] Open
Abstract
BackgroundMicrobial infection is accompanied by remodeling of the host transcriptome. Involvement of A-to-I RNA editing has been reported during viral infection but remains to be elucidated during intracellular bacterial infections.ResultsHerein we analyzed A-to-I RNA editing during intracellular bacterial infections based on 18 RNA-Seq datasets of 210 mouse samples involving 7 tissue types and 8 intracellular bacterial pathogens (IBPs), and identified a consensus signature of RNA editing for IBP infections, mainly involving neutrophil-mediated innate immunity and lipid metabolism. Further comparison of host RNA editing patterns revealed remarkable similarities between pneumonia caused by IBPs and single-strand RNA (ssRNA) viruses, such as altered editing enzyme expression, editing site numbers, and levels. In addition, functional enrichment analysis of genes with RNA editing highlighted that the Rab GTPase family played a common and vital role in the host immune response to IBP and ssRNA viral infections, which was indicated by the consistent up-regulated RNA editing of Ras-related protein Rab27a. Nevertheless, dramatic differences between IBP and viral infections were also observed, and clearly distinguished the two types of intracellular infections.ConclusionOur study showed transcriptome-wide host A-to-I RNA editing alteration during IBP and ssRNA viral infections. By identifying and comparing consensus signatures of host A-to-I RNA editing, our analysis implicates the importance of host A-to-I RNA editing during these infections and provides new insights into the diagnosis and treatment of infectious diseases.
Collapse
Affiliation(s)
- Zhi-Yuan Wei
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Zhi-Xin Wang
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Jia-Huan Li
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Yan-Shuo Wen
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Di Gao
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Shou-Yue Xia
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Yu-Ning Li
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Xu-Bin Pan
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Yan-Shan Liu
- Department of Pediatric Laboratory, Wuxi Children’s Hospital, Wuxi, Jiangsu, China
| | - Yun-Yun Jin
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
- *Correspondence: Jian-Huan Chen, ; Yun-Yun Jin,
| | - Jian-Huan Chen
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
- *Correspondence: Jian-Huan Chen, ; Yun-Yun Jin,
| |
Collapse
|
9
|
Shibata K, Motozono C, Nagae M, Shimizu T, Ishikawa E, Motooka D, Okuzaki D, Izumi Y, Takahashi M, Fujimori N, Wing JB, Hayano T, Asai Y, Bamba T, Ogawa Y, Furutani-Seiki M, Shirai M, Yamasaki S. Symbiotic bacteria-dependent expansion of MR1-reactive T cells causes autoimmunity in the absence of Bcl11b. Nat Commun 2022; 13:6948. [PMID: 36376329 PMCID: PMC9663695 DOI: 10.1038/s41467-022-34802-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
MHC class I-related protein 1 (MR1) is a metabolite-presenting molecule that restricts MR1-reactive T cells including mucosal-associated invariant T (MAIT) cells. In contrast to MAIT cells, the function of other MR1-restricted T cell subsets is largely unknown. Here, we report that mice in which a T cell-specific transcription factor, B-cell lymphoma/leukemia 11B (Bcl11b), was ablated in immature thymocytes (Bcl11b∆iThy mice) develop chronic inflammation. Bcl11b∆iThy mice lack conventional T cells and MAIT cells, whereas CD4+IL-18R+ αβ T cells expressing skewed Traj33 (Jα33)+ T cell receptors (TCR) accumulate in the periphery, which are necessary and sufficient for the pathogenesis. The disorders observed in Bcl11b∆iThy mice are ameliorated by MR1-deficiency, transfer of conventional T cells, or germ-free conditions. We further show the crystal structure of the TCR expressed by Traj33+ T cells expanded in Bcl11b∆iThy mice. Overall, we establish that MR1-reactive T cells have pathogenic potential.
Collapse
Affiliation(s)
- Kensuke Shibata
- grid.268397.10000 0001 0660 7960Department of Microbiology and Immunology, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan ,grid.177174.30000 0001 2242 4849Department of Ophthalmology, Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan ,grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Chihiro Motozono
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.274841.c0000 0001 0660 6749Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0871 Japan
| | - Masamichi Nagae
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Takashi Shimizu
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Eri Ishikawa
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Daisuke Motooka
- grid.136593.b0000 0004 0373 3971Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Daisuke Okuzaki
- grid.136593.b0000 0004 0373 3971Single Cell Genomics, Human Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Yoshihiro Izumi
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Masatomo Takahashi
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Nao Fujimori
- grid.177174.30000 0001 2242 4849Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - James B. Wing
- grid.136593.b0000 0004 0373 3971Laboratory of Human Immunology (Single Cell Immunology), World Premier International Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Takahide Hayano
- grid.268397.10000 0001 0660 7960Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Yoshiyuki Asai
- grid.268397.10000 0001 0660 7960Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Takeshi Bamba
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Yoshihiro Ogawa
- grid.177174.30000 0001 2242 4849Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan ,grid.419082.60000 0004 1754 9200Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology, Tokyo, 100-0004 Japan ,grid.27476.300000 0001 0943 978XDepartment of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601 Japan
| | - Makoto Furutani-Seiki
- grid.268397.10000 0001 0660 7960Systems Biochemistry in Pathology and Regeneration, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Mutsunori Shirai
- grid.268397.10000 0001 0660 7960Department of Microbiology and Immunology, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Sho Yamasaki
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.177174.30000 0001 2242 4849Division of Molecular Design, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan ,grid.136304.30000 0004 0370 1101Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, 260-8673 Japan
| |
Collapse
|
10
|
Oh SF, Jung DJ, Choi E. Gut Microbiota-Derived Unconventional T Cell Ligands: Contribution to Host Immune Modulation. Immunohorizons 2022; 6:476-487. [PMID: 35868838 PMCID: PMC9924074 DOI: 10.4049/immunohorizons.2200006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/28/2022] [Indexed: 01/26/2023] Open
Abstract
Besides the prototypic innate and adaptive pathways, immune responses by innate-like lymphocytes have gained significant attention due to their unique roles. Among innate-like lymphocytes, unconventional T cells such as NKT cells and mucosal-associated invariant T (MAIT) cells recognize small nonpeptide molecules of specific chemical classes. Endogenous or microbial ligands are loaded to MHC class I-like molecule CD1d or MR1, and inducing immediate effector T cell and ligand structure is one of the key determinants of NKT/MAIT cell functions. Unconventional T cells are in close, constant contact with symbiotic microbes at the mucosal layer, and CD1d/MR1 can accommodate diverse metabolites produced by gut microbiota. There is a strong interest to identify novel immunoactive molecules of endobiotic (symbiont-produced) origin as new NKT/MAIT cell ligands, as well as new cognate Ags for previously uncharacterized unconventional T cell subsets. Further studies will open an possibility to explore basic biology as well as therapeutic potential.
Collapse
Affiliation(s)
- Sungwhan F. Oh
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Da-Jung Jung
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Eungyo Choi
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
11
|
Morgan RC, Kee BL. Genomic and Transcriptional Mechanisms Governing Innate-like T Lymphocyte Development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:208-216. [PMID: 35821098 DOI: 10.4049/jimmunol.2200141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/18/2022] [Indexed: 12/16/2022]
Abstract
Innate-like lymphocytes are a subset of lymphoid cells that function as a first line of defense against microbial infection. These cells are activated by proinflammatory cytokines or broadly expressed receptors and are able to rapidly perform their effector functions owing to a uniquely primed chromatin state that is acquired as a part of their developmental program. These cells function in many organs to protect against disease, but they release cytokines and cytotoxic mediators that can also lead to severe tissue pathologies. Therefore, harnessing the capabilities of these cells for therapeutic interventions will require a deep understanding of how these cells develop and regulate their effector functions. In this review we discuss recent advances in the identification of the transcription factors and the genomic regions that guide the development and function of invariant NKT cells and we highlight related mechanisms in other innate-like lymphocytes.
Collapse
Affiliation(s)
- Roxroy C Morgan
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL; and
| | - Barbara L Kee
- Cancer Biology and Immunology, Department of Pathology, University of Chicago, Chicago, IL
| |
Collapse
|
12
|
Li Y, Du J, Wei W. Emerging Roles of Mucosal-Associated Invariant T Cells in Rheumatology. Front Immunol 2022; 13:819992. [PMID: 35317168 PMCID: PMC8934402 DOI: 10.3389/fimmu.2022.819992] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/14/2022] [Indexed: 12/22/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are an unconventional T cell subset expressing a semi-invariant TCR and recognize microbial riboflavin metabolites presented by major histocompatibility complex class 1-related molecule (MR1). MAIT cells serve as innate-like T cells bridging innate and adaptive immunity, which have attracted increasing attention in recent years. The involvement of MAIT cells has been described in various infections, autoimmune diseases and malignancies. In this review, we first briefly introduce the biology of MAIT cells, and then summarize their roles in rheumatic diseases including systemic lupus erythematosus, rheumatoid arthritis, primary Sjögren’s syndrome, psoriatic arthritis, systemic sclerosis, vasculitis and dermatomyositis. An increased knowledge of MAIT cells will inform the development of novel biomarkers and therapeutic approaches in rheumatology.
Collapse
|
13
|
Cheng ZY, He TT, Gao XM, Zhao Y, Wang J. ZBTB Transcription Factors: Key Regulators of the Development, Differentiation and Effector Function of T Cells. Front Immunol 2021; 12:713294. [PMID: 34349770 PMCID: PMC8326903 DOI: 10.3389/fimmu.2021.713294] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The development and differentiation of T cells represents a long and highly coordinated, yet flexible at some points, pathway, along which the sequential and dynamic expressions of different transcriptional factors play prominent roles at multiple steps. The large ZBTB family comprises a diverse group of transcriptional factors, and many of them have emerged as critical factors that regulate the lineage commitment, differentiation and effector function of hematopoietic-derived cells as well as a variety of other developmental events. Within the T-cell lineage, several ZBTB proteins, including ZBTB1, ZBTB17, ZBTB7B (THPOK) and BCL6 (ZBTB27), mainly regulate the development and/or differentiation of conventional CD4/CD8 αβ+ T cells, whereas ZBTB16 (PLZF) is essential for the development and function of innate-like unconventional γδ+ T & invariant NKT cells. Given the critical role of T cells in host defenses against infections/tumors and in the pathogenesis of many inflammatory disorders, we herein summarize the roles of fourteen ZBTB family members in the development, differentiation and effector function of both conventional and unconventional T cells as well as the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Zhong-Yan Cheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ting-Ting He
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathophysiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Jun Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
14
|
Lefferts AR, Regner EH, Stahly A, O'Rourke B, Gerich ME, Fennimore BP, Scott FI, Freeman AE, Jones K, Kuhn KA. Circulating mature granzyme B+ T cells distinguish Crohn's disease-associated axial spondyloarthritis from axial spondyloarthritis and Crohn's disease. Arthritis Res Ther 2021; 23:147. [PMID: 34022940 PMCID: PMC8140495 DOI: 10.1186/s13075-021-02531-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/12/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Axial spondyloarthritis (axSpA) has strong connections with intestinal inflammation as occurs in Crohn's disease (CD). However, the immunologic mechanisms that distinguish axSpA, CD, and those with features of both diseases (CD-axSpA) are unknown. This study aimed to address this question by initial unbiased single cell RNA-sequencing (scRNAseq) on a pilot cohort followed by validating findings using flow cytometry and ELISA in a larger cohort. METHODS Two individuals each with CD, axSpA, CD-axSpA, and healthy controls (HC) were recruited for a pilot discovery scRNAseq cohort, and the validation cohort consisted of 18 axSpA, 24 CD, 13 CD-axSpA, and 17 HC that was evaluated by flow cytometry on PBMCs and ELISAs for plasma cytokines. RESULTS Uniquely, PBMCs from subjects with CD-axSpA demonstrated a significant increase in granzyme B+ T cells of both CD4+ and CD8+ lineages by both scRNAseq and flow cytometry. T cell maturation was also greater in those with CD-axSpA, particularly the CD4+ granzyme B+ population. Pathway analysis suggested increased interferon response genes in all immune cell populations within CD-axSpA. Although IFN-γ was elevated in the plasma of a subset of subjects with CD-axSpA, IL-6 was also significantly elevated. CONCLUSIONS Our findings support the presence of a chronic interferonopathy in subjects with CD-axSpA characterized by interferon signaling by pathway analysis and an expansion of mature, cytotoxic T cells. These data indicate fundamental immunological differences between CD-axSpA and both of the putative "parent" conditions, suggesting that it is a distinct disease with unique natural history and treatment needs.
Collapse
Affiliation(s)
- Adam R Lefferts
- Division of Rheumatology, Department of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Emilie H Regner
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Present Address: Division of Gastroenterology, Department of Medicine, Oregon Health Sciences University, Portland, OR, USA
| | - Andrew Stahly
- Division of Rheumatology, Department of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Becky O'Rourke
- Section of Pediatric Hematology/Oncology/Bone Marrow Transplant, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Mark E Gerich
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Blair P Fennimore
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Frank I Scott
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Alison E Freeman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Present Address: Cascade Gastroenterology, Bend, OR, USA
| | - Ken Jones
- Section of Pediatric Hematology/Oncology/Bone Marrow Transplant, Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Present Address: Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kristine A Kuhn
- Division of Rheumatology, Department of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|