1
|
Jian N, Yu L, Ma L, Zheng B, Huang W. BCG therapy in bladder cancer and its tumor microenvironment interactions. Clin Microbiol Rev 2025:e0021224. [PMID: 40111053 DOI: 10.1128/cmr.00212-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
SUMMARYBacillus Calmette-Guérin (BCG) has been the standard treatment for non-muscle-invasive bladder cancer for over 30 years. Despite its proven efficacy, challenges persist, including unclear mechanisms of action, resistance in 30%-50% of patients, and significant side effects. This review presents an updated and balanced discussion of the antitumor mechanisms of BCG, focusing on its direct effects on bladder cancer and its interactions with various cell types within the bladder tumor microenvironment. Notably, recent research on the interactions between BCG and the bladder microbiome is also incorporated. We further summarize and analyze the latest preclinical and clinical studies regarding both intrinsic and adaptive resistance to BCG in bladder cancer. Based on the current understanding of BCG's therapeutic principles and resistance mechanisms, we systematically explore strategies to improve BCG-based tumor immunotherapy. These include the development of recombinant BCG, combination therapy with different drugs, optimization of therapeutic regimens and management, and the exploration of new approaches by targeting changes in the bladder microbiota and its metabolites. These measures aim to effectively address the BCG resistance in bladder cancer, reduce its toxicity, and ultimately enhance the clinical anti-tumor efficacy. Bacterial therapy, represented by genetically engineered oncolytic bacteria, has gradually emerged in the field of cancer treatment in recent years. As the only bacterial drug successfully approved for oncology use, BCG has provided decades of clinical experience. By consolidating lessons from BCG's successes and limitations, we hope to provide valuable insights for the development and application of bacterial therapies in cancer treatment.
Collapse
Affiliation(s)
- Ni Jian
- Synthetic Biology Research Center, Institute for Advanced Study, International Cancer Center of Shenzhen University, Shenzhen, China
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Lei Yu
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lijuan Ma
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Binbin Zheng
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Weiren Huang
- Synthetic Biology Research Center, Institute for Advanced Study, International Cancer Center of Shenzhen University, Shenzhen, China
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
2
|
Irla M. Thymic Crosstalk: An Overview of the Complex Cellular Interactions that Control the Establishment of T-Cell Tolerance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:177-197. [PMID: 40067587 DOI: 10.1007/978-3-031-77921-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The thymus ensures the generation of a self-tolerant T-cell repertoire capable of recognizing foreign antigens. The selection of the T-cell repertoire is dictated by the thymic microenvironment. Among stromal cells, medullary thymic epithelial cells (mTECs) play a pivotal role in this process through their unique ability to express thousands of tissue-restricted self-antigens. In turn, developing T cells control the pool and maturation of mTECs. This phenomenon of bidirectional interactions between TECs and thymocytes is referred to as thymic crosstalk. In this chapter, I discuss the discovery of thymic crosstalk and our current understanding of bidirectional interactions between mTECs and thymocytes. Finally, I summarize recent advances indicating that thymic crosstalk is not restricted to TECs and thymocytes but also occurs between TECs and dendritic cells, as well as B cells and thymocytes. This complex cellular interplay is essential for efficient T-cell selection.
Collapse
Affiliation(s)
- Magali Irla
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| |
Collapse
|
3
|
Rane G, Kuan VLS, Wang S, Mok MMH, Khanchandani V, Hansen J, Norvaisaite I, Zulkaflee N, Yong WK, Jahn A, Mukundan VT, Shi Y, Osato M, Li F, Kappei D. ZBTB48 is a priming factor regulating B-cell-specific CIITA expression. EMBO J 2024; 43:6236-6263. [PMID: 39562739 PMCID: PMC11649694 DOI: 10.1038/s44318-024-00306-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024] Open
Abstract
The class-II transactivator (CIITA) is the master regulator of MHC class-II gene expression and hence the adaptive immune response. Three cell type-specific promoters (pI, pIII, and pIV) are involved in the regulation of CIITA expression, which can be induced by IFN-γ in non-immune cells. While key regulatory elements have been identified within these promoters, our understanding of the transcription factors regulating CIITA expression is incomplete. Here, we demonstrate that the telomere-binding protein and transcriptional activator ZBTB48 directly binds to both critical activating elements within the B-cell-specific promoter CIITA pIII. ZBTB48 knockout impedes the CIITA/MHC-II expression program induced in non-APC cells by IFN-γ, and loss of ZBTB48 in mice silences MHC-II expression in pro-B and immature B cells. Transcriptional regulation of CIITA by ZBTB48 is enabled by ZBTB48-dependent chromatin opening at CIITA pIII upstream of activating H3K4me3 marks. We conclude that ZBTB48 primes CIITA pIII by acting as a molecular on-off-switch for B-cell-specific CIITA expression.
Collapse
Affiliation(s)
- Grishma Rane
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Vivian L S Kuan
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Suman Wang
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Michelle Meng Huang Mok
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Vartika Khanchandani
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Julia Hansen
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Ieva Norvaisaite
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Naasyidah Zulkaflee
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Wai Khang Yong
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Arne Jahn
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- ERN-GENTURIS, Hereditary Cancer Syndrome Center, Dresden, Germany
| | - Vineeth T Mukundan
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Yunyu Shi
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Motomi Osato
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - Fudong Li
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596, Singapore, Singapore.
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Chang YH, Yamamoto K, Fujino T, Wang TW, Sugimoto E, Zhang W, Yabushita T, Suzaki K, Pietsch EC, Weir BA, Crescenzo R, Cowley GS, Attar R, Philippar U, Wunderlich M, Mizukawa B, Zheng Y, Enomoto Y, Imai Y, Kitamura T, Goyama S. SETDB1 suppresses NK cell-mediated immunosurveillance in acute myeloid leukemia with granulo-monocytic differentiation. Cell Rep 2024; 43:114536. [PMID: 39096901 DOI: 10.1016/j.celrep.2024.114536] [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: 11/17/2023] [Revised: 05/15/2024] [Accepted: 07/09/2024] [Indexed: 08/05/2024] Open
Abstract
Monocytic acute myeloid leukemia (AML) responds poorly to current treatments, including venetoclax-based therapy. We conducted in vivo and in vitro CRISPR-Cas9 library screenings using a mouse monocytic AML model and identified SETDB1 and its binding partners (ATF7IP and TRIM33) as crucial tumor promoters in vivo. The growth-inhibitory effect of Setdb1 depletion in vivo is dependent mainly on natural killer (NK) cell-mediated cytotoxicity. Mechanistically, SETDB1 depletion upregulates interferon-stimulated genes and NKG2D ligands through the demethylation of histone H3 Lys9 at the enhancer regions, thereby enhancing their immunogenicity to NK cells and intrinsic apoptosis. Importantly, these effects are not observed in non-monocytic leukemia cells. We also identified the expression of myeloid cell nuclear differentiation antigen (MNDA) and its murine counterpart Ifi203 as biomarkers to predict the sensitivity of AML to SETDB1 depletion. Our study highlights the critical and selective role of SETDB1 in AML with granulo-monocytic differentiation and underscores its potential as a therapeutic target for current unmet needs.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Animals
- Cell Differentiation
- Mice
- Histone-Lysine N-Methyltransferase/metabolism
- Histone-Lysine N-Methyltransferase/genetics
- Humans
- Mice, Inbred C57BL
- Cell Line, Tumor
- Immunologic Surveillance
- Monocytes/metabolism
- Monocytes/immunology
- Apoptosis
Collapse
Affiliation(s)
- Yu-Hsuan Chang
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan; Division of Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-8654, Japan
| | - Keita Yamamoto
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
| | - Takeshi Fujino
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
| | - Teh-Wei Wang
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Emi Sugimoto
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
| | - Wenyu Zhang
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tomohiro Yabushita
- Division of Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-8654, Japan
| | - Ken Suzaki
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
| | | | - Barbara A Weir
- Janssen Research and Development, Cambridge, MA 02141, USA
| | | | - Glenn S Cowley
- Janssen Research and Development, Spring House, PA 19002, USA
| | - Ricardo Attar
- Janssen Research and Development, Spring House, PA 19002, USA
| | | | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Benjamin Mizukawa
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yutaka Enomoto
- Division of Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-8654, Japan
| | - Yoichi Imai
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Toshio Kitamura
- Division of Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-8654, Japan; Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan
| | - Susumu Goyama
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan.
| |
Collapse
|
5
|
Burdett NL, Willis MO, Pandey A, Twomey L, Alaei S, Bowtell DDL, Christie EL. Timing of whole genome duplication is associated with tumor-specific MHC-II depletion in serous ovarian cancer. Nat Commun 2024; 15:6069. [PMID: 39025846 PMCID: PMC11258338 DOI: 10.1038/s41467-024-50137-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
Whole genome duplication is frequently observed in cancer, and its prevalence in our prior analysis of end-stage, homologous recombination deficient high grade serous ovarian cancer (almost 80% of samples) supports the notion that whole genome duplication provides a fitness advantage under the selection pressure of therapy. Here, we therefore aim to identify potential therapeutic vulnerabilities in primary high grade serous ovarian cancer with whole genome duplication by assessing differentially expressed genes and pathways in 79 samples. We observe that MHC-II expression is lowest in tumors which have acquired whole genome duplication early in tumor evolution, and further demonstrate that reduced MHC-II expression occurs in subsets of tumor cells rather than in canonical antigen-presenting cells. Early whole genome duplication is also associated with worse patient survival outcomes. Our results suggest an association between the timing of whole genome duplication, MHC-II expression and clinical outcome in high grade serous ovarian cancer that warrants further investigation for therapeutic targeting.
Collapse
Affiliation(s)
- Nikki L Burdett
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Box Hill Hospital, Eastern Health, Box Hill, VIC, 3128, Australia
| | | | - Ahwan Pandey
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Laura Twomey
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Sara Alaei
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3168, Australia
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
6
|
Clements RL, Kennedy EA, Song D, Campbell A, An HH, Amses KR, Miller-Ensminger T, Addison MM, Eisenlohr LC, Chou ST, Jurado KA. Human erythroid progenitors express antigen presentation machinery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601047. [PMID: 39005276 PMCID: PMC11244935 DOI: 10.1101/2024.06.27.601047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Early-life immune exposures can profoundly impact lifelong health. However, functional mechanisms underlying fetal immune development remain incomplete. Erythrocytes are not typically considered active immune mediators, primarily because erythroid precursors discard their organelles as they mature, thus losing the ability to alter gene expression in response to stimuli. Erythroid progenitors and precursors circulate in human fetuses and neonates. Although there is limited evidence that erythroid precursors are immunomodulatory, our understanding of the underlying mechanisms remains inadequate. To define the immunobiological role of fetal and perinatal erythroid progenitors and precursors, we analyzed single cell RNA-sequencing data and found that transcriptomics support erythroid progenitors as putative immune mediators. Unexpectedly, we discovered that human erythroid progenitors constitutively express Major Histocompatibility Complex (MHC) class II antigen processing and presentation machinery, which are hallmarks of specialized antigen presenting immune cells. Furthermore, we demonstrate that erythroid progenitors internalize and cleave foreign proteins into peptide antigens. Unlike conventional antigen presenting cells, erythroid progenitors express atypical costimulatory molecules and immunoregulatory cytokines that direct the development of regulatory T cells, which are critical for establishing maternal-fetal tolerance. Expression of MHC II in definitive erythroid progenitors begins during the second trimester, coinciding with the appearance of mature T cells in the fetus, and is absent in primitive progenitors. Lastly, we demonstrate physical and molecular interaction potential of erythroid progenitors and T cells in the fetal liver. Our findings shed light on a unique orchestrator of fetal immunity and provide insight into the mechanisms by which erythroid cells contribute to host defense.
Collapse
|
7
|
Heuberger CE, Janney A, Ilott N, Bertocchi A, Pott S, Gu Y, Pohin M, Friedrich M, Mann EH, Pearson C, Powrie FM, Pott J, Thornton E, Maloy KJ. MHC class II antigen presentation by intestinal epithelial cells fine-tunes bacteria-reactive CD4 T-cell responses. Mucosal Immunol 2024; 17:416-430. [PMID: 37209960 DOI: 10.1016/j.mucimm.2023.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/18/2023] [Accepted: 05/01/2023] [Indexed: 05/22/2023]
Abstract
Although intestinal epithelial cells (IECs) can express major histocompatibility complex class II (MHC II), especially during intestinal inflammation, it remains unclear if antigen presentation by IECs favors pro- or anti-inflammatory CD4+ T-cell responses. Using selective gene ablation of MHC II in IECs and IEC organoid cultures, we assessed the impact of MHC II expression by IECs on CD4+ T-cell responses and disease outcomes in response to enteric bacterial pathogens. We found that intestinal bacterial infections elicit inflammatory cues that greatly increase expression of MHC II processing and presentation molecules in colonic IECs. Whilst IEC MHC II expression had little impact on disease severity following Citrobacter rodentium or Helicobacter hepaticus infection, using a colonic IEC organoid-CD4+ T cell co-culture system, we demonstrate that IECs can activate antigen-specific CD4+ T cells in an MHC II-dependent manner, modulating both regulatory and effector Th cell subsets. Furthermore, we assessed adoptively transferred H. hepaticus-specific CD4+ T cells during intestinal inflammation in vivo and report that IEC MHC II expression dampens pro-inflammatory effector Th cells. Our findings indicate that IECs can function as non-conventional antigen-presenting cells and that IEC MHC II expression fine-tunes local effector CD4+ T-cell responses during intestinal inflammation.
Collapse
Affiliation(s)
- Cornelia E Heuberger
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom; Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Alina Janney
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Nicholas Ilott
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Alice Bertocchi
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Sebastian Pott
- Department of Human Genetics, University of Chicago, Chicago, United States
| | - Yisu Gu
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Mathilde Pohin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Matthias Friedrich
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom; Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Elizabeth H Mann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Claire Pearson
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Fiona M Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Johanna Pott
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom; Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Emily Thornton
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Kevin Joseph Maloy
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
8
|
Pitter MR, Kryczek I, Zhang H, Nagarsheth N, Xia H, Wu Z, Tian Y, Okla K, Liao P, Wang W, Zhou J, Li G, Lin H, Vatan L, Grove S, Wei S, Li Y, Zou W. PAD4 controls tumor immunity via restraining the MHC class II machinery in macrophages. Cell Rep 2024; 43:113942. [PMID: 38489266 PMCID: PMC11022165 DOI: 10.1016/j.celrep.2024.113942] [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: 09/08/2023] [Revised: 01/18/2024] [Accepted: 02/26/2024] [Indexed: 03/17/2024] Open
Abstract
Tumor-associated macrophages (TAMs) shape tumor immunity and therapeutic efficacy. However, it is poorly understood whether and how post-translational modifications (PTMs) intrinsically affect the phenotype and function of TAMs. Here, we reveal that peptidylarginine deiminase 4 (PAD4) exhibits the highest expression among common PTM enzymes in TAMs and negatively correlates with the clinical response to immune checkpoint blockade. Genetic and pharmacological inhibition of PAD4 in macrophages prevents tumor progression in tumor-bearing mouse models, accompanied by an increase in macrophage major histocompatibility complex (MHC) class II expression and T cell effector function. Mechanistically, PAD4 citrullinates STAT1 at arginine 121, thereby promoting the interaction between STAT1 and protein inhibitor of activated STAT1 (PIAS1), and the loss of PAD4 abolishes this interaction, ablating the inhibitory role of PIAS1 in the expression of MHC class II machinery in macrophages and enhancing T cell activation. Thus, the PAD4-STAT1-PIAS1 axis is an immune restriction mechanism in macrophages and may serve as a cancer immunotherapy target.
Collapse
Affiliation(s)
- Michael R Pitter
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Graduate Program in Molecular and Cellular Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Hongjuan Zhang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Nisha Nagarsheth
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Houjun Xia
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Zhenyu Wu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yuzi Tian
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Karolina Okla
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Peng Liao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Weichao Wang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Jiajia Zhou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Gaopeng Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Heng Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Yongqing Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Graduate Programs in Immunology and Cancer Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Tsankov BK, Luchak A, Carr C, Philpott DJ. The effects of NOD-like receptors on adaptive immune responses. Biomed J 2024; 47:100637. [PMID: 37541620 PMCID: PMC10796267 DOI: 10.1016/j.bj.2023.100637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023] Open
Abstract
It has long been appreciated that cues from the innate immune system orchestrate downstream adaptive immune responses. Although previous work has focused on the roles of Toll-like receptors in this regard, relatively little is known about how Nod-like receptors instruct adaptive immunity. Here we review the functions of different members of the Nod-like receptor family in orchestrating effector and anamnestic adaptive immune responses. In particular, we address the ways in which inflammasome and non-inflammasome members of this family affect adaptive immunity under various infectious and environmental contexts. Furthermore, we identify several key mechanistic questions that studies in this field have left unaddressed. Our aim is to provide a framework through which immunologists in the adaptive immune field may view their questions through an innate-immune lens and vice-versa.
Collapse
Affiliation(s)
- Boyan K Tsankov
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Alexander Luchak
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Charles Carr
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Han Y, Zhang X, Wang Q, Cui X, Wang H, Zhang X, Wang Q, Ji J, Wang Y, Wang S, Zhang X, Xu H, Qiao M, Wu Z. IL-27p28 specifically regulates MHC II expression in macrophages through CIITA. Immunobiology 2023; 228:152757. [PMID: 37944428 DOI: 10.1016/j.imbio.2023.152757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/08/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Antigen-presenting cells (APCs) constantly express major histocompatibility complex II (MHC II), including macrophages and dendritic cells (DCs) which deliver antigens to CD4+ T cells and play an important role in adaptive immunity. The expression of MHC II is controlled by the transcriptional coactivator CIITA. Interleukin-27 (IL-27), a newly discovered IL-12 family cytokine, is composed of p28 and EBI3 subunits. In this study, we used IL-27p28 conditional knock-out mice to investigate the regulatory effects of IL-27p28 on macrophage polarization and the expression of MHC II in macrophages. We found that MHC II expression was upregulated in the bone marrow-derived and peritoneal exudate macrophages (BMDMs; PEMs) from IL-27p28-deficient mice, with their inflammation regulating function unaffected. We also demonstrated that in the APCs, IL-27p28 selectively regulated MHC II expression in macrophages but not in dendritic cells. During Pseudomonas aeruginosa (P. aeruginosa) reinfection, higher survival rate, bacterial clearance, and ratio of CD4+/CD8+ T cells in the spleen during the specific immune phase were observed in IL-27p28 defect mice, as well as an increased MHC II expression in alveolar macrophages (AMs). But these did not occur in the first infection. For the first time we discovered that IL-27p28 specifically regulates the expression of MHC II in macrophages by regulating CIITA, while its absence enhances antigen presentation and adaptive immunity against P. aeruginosa.
Collapse
Affiliation(s)
- Yu Han
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xu Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Qing Wang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoyue Cui
- College of Life Sciences, Nankai University, Tianjin, China
| | - Hesuiyuan Wang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiang Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Qian Wang
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Jianbin Ji
- First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuebing Wang
- School of Medicine, Nankai University, Tianjin, China
| | - Shusen Wang
- Tianjin First Central Hospital, Tianjin, China
| | - Xiuming Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Haijin Xu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Mingqiang Qiao
- College of Life Sciences, Nankai University, Tianjin, China
| | - Zhenzhou Wu
- College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
11
|
Redelman-Sidi G, Binyamin A, Antonelli AC, Catalano W, Bean J, Al-Ahmadie H, Jungbluth AA, Glickman MS. BCG-Induced Tumor Immunity Requires Tumor-Intrinsic CIITA Independent of MHC-II. Cancer Immunol Res 2022; 10:1241-1253. [PMID: 36040405 PMCID: PMC9532361 DOI: 10.1158/2326-6066.cir-22-0157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/09/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022]
Abstract
For decades, BCG immunotherapy has been the standard of care for non-muscle-invasive bladder cancer. Despite this clinical experience, the mechanism by which BCG stimulates tumor-eliminating immunity is unclear, and there is still a need for more accurate prediction of clinical outcomes in advance of treatment initiation. We have shown that BCG stimulates tumor-specific T-cell immunity that requires tumor cell expression of the IFNγ receptor (IFNGR); however, the downstream components of IFNGR signaling responsible for responsiveness to BCG are unknown. Here, we demonstrate that the IFNγ-driven, tumor cell intrinsic expression of the class II transactivator CIITA is required for activation of a tumor-specific CD4 T-cell response and BCG-induced tumor immunity. Despite the established role for CIITA in controlling MHC-II antigen presentation machinery, the requirement for CIITA is independent of MHC-II and associated genes. Rather, we find that CIITA is required for a broader tumor-intrinsic transcriptional program linked to critical pathways of tumor immunity via mechanisms that remain to be determined. Tumor cell intrinsic expression of CIITA is not required for a response to immunotherapy targeting programmed cell death protein 1 (PD-1), suggesting that different modalities of immunotherapy for bladder cancer could be employed based on tumor-intrinsic characteristics.
Collapse
Affiliation(s)
- Gil Redelman-Sidi
- Division of Infectious Diseases, Memorial Sloan Kettering Cancer Center
| | | | - Anthony C. Antonelli
- Immunology Program, Sloan Kettering Institute
- Immunology and Microbial Pathogenesis, Weill Cornell Medicine Graduate School of Medical Sciences
| | | | - James Bean
- Immunology Program, Sloan Kettering Institute
| | | | | | - Michael S. Glickman
- Division of Infectious Diseases, Memorial Sloan Kettering Cancer Center
- Immunology Program, Sloan Kettering Institute
| |
Collapse
|
12
|
Toulmin SA, Bhadiadra C, Paris AJ, Lin JH, Katzen J, Basil MC, Morrisey EE, Worthen GS, Eisenlohr LC. Type II alveolar cell MHCII improves respiratory viral disease outcomes while exhibiting limited antigen presentation. Nat Commun 2021; 12:3993. [PMID: 34183650 PMCID: PMC8239023 DOI: 10.1038/s41467-021-23619-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 05/05/2021] [Indexed: 02/06/2023] Open
Abstract
Type II alveolar cells (AT2s) are critical for basic respiratory homeostasis and tissue repair after lung injury. Prior studies indicate that AT2s also express major histocompatibility complex class II (MHCII) molecules, but how MHCII expression by AT2s is regulated and how it contributes to host defense remain unclear. Here we show that AT2s express high levels of MHCII independent of conventional inflammatory stimuli, and that selective loss of MHCII from AT2s in mice results in modest worsening of respiratory virus disease following influenza and Sendai virus infections. We also find that AT2s exhibit MHCII presentation capacity that is substantially limited compared to professional antigen presenting cells. The combination of constitutive MHCII expression and restrained antigen presentation may position AT2s to contribute to lung adaptive immune responses in a measured fashion, without over-amplifying damaging inflammation.
Collapse
Affiliation(s)
- Sushila A. Toulmin
- grid.239552.a0000 0001 0680 8770Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Chaitali Bhadiadra
- grid.239552.a0000 0001 0680 8770Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Andrew J. Paris
- grid.25879.310000 0004 1936 8972Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Jeffrey H. Lin
- grid.25879.310000 0004 1936 8972Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Jeremy Katzen
- grid.25879.310000 0004 1936 8972Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Maria C. Basil
- grid.25879.310000 0004 1936 8972Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA USA
| | - Edward E. Morrisey
- grid.25879.310000 0004 1936 8972Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Penn Institute for Regenerative Medicine, Perelman School of Medicine, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - G. Scott Worthen
- grid.25879.310000 0004 1936 8972Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA ,grid.239552.a0000 0001 0680 8770Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Laurence C. Eisenlohr
- grid.239552.a0000 0001 0680 8770Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
13
|
Heuberger C, Pott J, Maloy KJ. Why do intestinal epithelial cells express MHC class II? Immunology 2021; 162:357-367. [PMID: 32966619 PMCID: PMC7968399 DOI: 10.1111/imm.13270] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/23/2022] Open
Abstract
Intestinal epithelial cells (IECs) constitute the border between the vast antigen load present in the intestinal lumen and the mucosal immune compartment. Their ability to express antigen processing and presentation machinery evokes the question whether IECs function as non-conventional antigen-presenting cells. Major histocompatibility complex (MHC) class II expression by non-haematopoietic cells, such as IECs, is tightly regulated by the class II transactivator (CIITA) and is classically induced by IFN-γ. As MHC class II expression by IECs is upregulated under inflammatory conditions, it has been proposed to activate effector CD4+ T (Teff) cells. However, other studies have reported contradictory results and instead suggested a suppressive role of antigen presentation by IECs, through regulatory T (Treg)-cell activation. Recent studies investigating the role of MHC class II + exosomes released by IECs also reported conflicting findings of either immune enhancing or immunosuppressive activities. Moreover, in addition to modulating inflammatory responses, recent findings suggest that MHC class II expression by intestinal stem cells may elicit crosstalk that promotes epithelial renewal. A more complete understanding of the different consequences of IEC MHC class II antigen presentation will guide future efforts to modulate this pathway to selectively invoke protective immunity while maintaining tolerance to beneficial antigens.
Collapse
Affiliation(s)
- Cornelia Heuberger
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Johanna Pott
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Hubrecht Organoid TechnologyUtrechtNetherlands
| | - Kevin Joseph Maloy
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Institute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| |
Collapse
|
14
|
León Machado JA, Steimle V. The MHC Class II Transactivator CIITA: Not (Quite) the Odd-One-Out Anymore among NLR Proteins. Int J Mol Sci 2021; 22:1074. [PMID: 33499042 PMCID: PMC7866136 DOI: 10.3390/ijms22031074] [Citation(s) in RCA: 36] [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: 12/23/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
In this review, we discuss the major histocompatibility complex (MHC) class II transactivator (CIITA), which is the master regulator of MHC class II gene expression. CIITA is the founding member of the mammalian nucleotide-binding and leucine-rich-repeat (NLR) protein family but stood apart for a long time as the only transcriptional regulator. More recently, it was found that its closest homolog, NLRC5 (NLR protein caspase activation and recruitment domain (CARD)-containing 5), is a regulator of MHC-I gene expression. Both act as non-DNA-binding activators through multiple protein-protein interactions with an MHC enhanceosome complex that binds cooperatively to a highly conserved combinatorial cis-acting module. Thus, the regulation of MHC-II expression is regulated largely through the differential expression of CIITA. In addition to the well-defined role of CIITA in MHC-II GENE regulation, we will discuss several other aspects of CIITA functions, such as its role in cancer, its role as a viral restriction element contributing to intrinsic immunity, and lastly, its very recently discovered role as an inhibitor of Ebola and SARS-Cov-2 virus replication. We will briefly touch upon the recently discovered role of NLRP3 as a transcriptional regulator, which suggests that transcriptional regulation is, after all, not such an unusual feature for NLR proteins.
Collapse
Affiliation(s)
| | - Viktor Steimle
- Département de Biologie, Université de Sherbrooke, 2500 Boul., Sherbrooke, QC J1K 2R1, Canada;
| |
Collapse
|
15
|
Biffi G, Tuveson DA. Diversity and Biology of Cancer-Associated Fibroblasts. Physiol Rev 2021; 101:147-176. [PMID: 32466724 PMCID: PMC7864232 DOI: 10.1152/physrev.00048.2019] [Citation(s) in RCA: 720] [Impact Index Per Article: 180.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023] Open
Abstract
Efforts to develop anti-cancer therapies have largely focused on targeting the epithelial compartment, despite the presence of non-neoplastic stromal components that substantially contribute to the progression of the tumor. Indeed, cancer cell survival, growth, migration, and even dormancy are influenced by the surrounding tumor microenvironment (TME). Within the TME, cancer-associated fibroblasts (CAFs) have been shown to play several roles in the development of a tumor. They secrete growth factors, inflammatory ligands, and extracellular matrix proteins that promote cancer cell proliferation, therapy resistance, and immune exclusion. However, recent work indicates that CAFs may also restrain tumor progression in some circumstances. In this review, we summarize the body of work on CAFs, with a particular focus on the most recent discoveries about fibroblast heterogeneity, plasticity, and functions. We also highlight the commonalities of fibroblasts present across different cancer types, and in normal and inflammatory states. Finally, we present the latest advances regarding therapeutic strategies targeting CAFs that are undergoing preclinical and clinical evaluation.
Collapse
Affiliation(s)
- Giulia Biffi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York; and Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York; and Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
16
|
Brouwer MAE, Jones-Warner W, Rahman S, Kerstholt M, Ferreira AV, Oosting M, Hooiveld GJ, Netea MG, Joosten LAB. B. burgdorferi sensu lato-induced inhibition of antigen presentation is mediated by RIP1 signaling resulting in impaired functional T cell responses towards Candida albicans. Ticks Tick Borne Dis 2020; 12:101611. [PMID: 33360386 DOI: 10.1016/j.ttbdis.2020.101611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
Antigen presentation is a crucial innate immune cell function that instructs adaptive immune cells. Loss of this pathway severely impairs the development of adaptive immune responses. To investigate whether B. burgdorferi sensu lato. spirochetes modulate the induction of an effective immune response, primary human PBMCs were isolated from healthy volunteers and stimulated with B. burgdorferi s.l. Through cell entry, TNF receptor I, and RIP1 signaling cascades, B. burgdorferi s.l. strongly downregulated genes and proteins involved in antigen presentation, specifically HLA-DM, MHC class II and CD74. Antigen presentation proteins were distinctively inhibited in monocyte subsets, monocyte-derived macrophages, and dendritic cells. When compared to a range of other pathogens, B. burgdorferi s.l.-induced suppression of antigen presentation appears to be specific. Inhibition of antigen presentation interfered with T-cell recognition of B. burgdorferi s.l., and memory T-cell responses against Candidaalbicans. Re-stimulation of PBMCs with the commensal microbe C.albicans following B. burgdorferi s.l. exposure resulted in significantly reduced IFN-γ, IL-17 and IL-22 production. These findings may explain why patients with Lyme borreliosis develop delayed adaptive immune responses. Unravelling the mechanism of B. burgdorferi s.l.-induced inhibition of antigen presentation, via cell entry, TNF receptor I, and RIP1 signaling cascades, explains the difficulty to diagnose the disease based on serology and to obtain an effective vaccine against Lyme borreliosis.
Collapse
Affiliation(s)
- Michelle A E Brouwer
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - William Jones-Warner
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Shafaque Rahman
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Mariska Kerstholt
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Anaísa V Ferreira
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Marije Oosting
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Guido J Hooiveld
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
17
|
Lymph Node Stromal Cells: Mapmakers of T Cell Immunity. Int J Mol Sci 2020; 21:ijms21207785. [PMID: 33096748 PMCID: PMC7588999 DOI: 10.3390/ijms21207785] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022] Open
Abstract
Stromal cells (SCs) are strategically positioned in both lymphoid and nonlymphoid organs to provide a scaffold and orchestrate immunity by modulating immune cell maturation, migration and activation. Recent characterizations of SCs have expanded our understanding of their heterogeneity and suggested a functional specialization of distinct SC subsets, further modulated by the microenvironment. Lymph node SCs (LNSCs) have been shown to be particularly important in maintaining immune homeostasis and T cell tolerance. Under inflammation situations, such as viral infections or tumor development, SCs undergo profound changes in their numbers and phenotype and play important roles in contributing to either the activation or the control of T cell immunity. In this review, we highlight the role of SCs located in LNs in shaping peripheral T cell responses in different immune contexts, such as autoimmunity, viral and cancer immunity.
Collapse
|
18
|
Friedman G, Levi-Galibov O, David E, Bornstein C, Giladi A, Dadiani M, Mayo A, Halperin C, Pevsner-Fischer M, Lavon H, Mayer S, Nevo R, Stein Y, Balint-Lahat N, Barshack I, Ali HR, Caldas C, Nili-Gal-Yam E, Alon U, Amit I, Scherz-Shouval R. Cancer-associated fibroblast compositions change with breast cancer progression linking the ratio of S100A4 + and PDPN + CAFs to clinical outcome. NATURE CANCER 2020; 1:692-708. [PMID: 35122040 PMCID: PMC7617059 DOI: 10.1038/s43018-020-0082-y] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/19/2020] [Indexed: 02/01/2023]
Abstract
Tumors are supported by cancer-associated fibroblasts (CAFs). CAFs are heterogeneous and carry out distinct cancer-associated functions. Understanding the full repertoire of CAFs and their dynamic changes as tumors evolve could improve the precision of cancer treatment. Here we comprehensively analyze CAFs using index and transcriptional single-cell sorting at several time points along breast tumor progression in mice, uncovering distinct subpopulations. Notably, the transcriptional programs of these subpopulations change over time and in metastases, transitioning from an immunoregulatory program to wound-healing and antigen-presentation programs, indicating that CAFs and their functions are dynamic. Two main CAF subpopulations are also found in human breast tumors, where their ratio is associated with disease outcome across subtypes and is particularly correlated with BRCA mutations in triple-negative breast cancer. These findings indicate that the repertoire of CAF changes over time in breast cancer progression, with direct clinical implications.
Collapse
Affiliation(s)
- Gil Friedman
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Oshrat Levi-Galibov
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Eyal David
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Chamutal Bornstein
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Amir Giladi
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Maya Dadiani
- Chaim Sheba Medical Center, Cancer Research Center, Tel-Hashomer, Israel
| | - Avi Mayo
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Coral Halperin
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | | | - Hagar Lavon
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Shimrit Mayer
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Reinat Nevo
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Yaniv Stein
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | | | - Iris Barshack
- Pathology Institute, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - H Raza Ali
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
- Breast Cancer Programme, Cancer Research UK Cancer Centre, Cambridge, UK
| | | | - Uri Alon
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ido Amit
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
19
|
Hou J, Chen SN, Gan Z, Li N, Huang L, Huo HJ, Yang YC, Lu Y, Yin Z, Nie P. In Primitive Zebrafish, MHC Class II Expression Is Regulated by IFN-γ, IRF1, and Two Forms of CIITA. THE JOURNAL OF IMMUNOLOGY 2020; 204:2401-2415. [DOI: 10.4049/jimmunol.1801480] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/13/2020] [Indexed: 12/21/2022]
|
20
|
Lochhead RB, Ordoñez D, Arvikar SL, Aversa JM, Oh LS, Heyworth B, Sadreyev R, Steere AC, Strle K. Interferon-gamma production in Lyme arthritis synovial tissue promotes differentiation of fibroblast-like synoviocytes into immune effector cells. Cell Microbiol 2019; 21:e12992. [PMID: 30550623 DOI: 10.1111/cmi.12992] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 12/03/2018] [Accepted: 12/09/2018] [Indexed: 12/21/2022]
Abstract
Lyme arthritis (LA), a late disease manifestation of Borrelia burgdorferi infection, usually resolves with antibiotic therapy. However, some patients develop proliferative synovitis lasting months to several years after spirochetal killing, called postinfectious LA. In this study, we phenotyped haematopoietic and stromal cell populations in the synovial lesion ex vivo and used these findings to generate an in vitro model of LA using patient-derived fibroblast-like synoviocytes (FLS). Ex vivo analysis of synovial tissue revealed high abundance of IFNγ-producing T cells and NK cells. Similar to marked IFNγ responses in tissue, postinfectious LA synovial fluid also had high levels of IFNγ. HLA-DR-positive FLS were present throughout the synovial lesion, particularly in areas of inflammation. FLS stimulated in vitro with B. burgdorferi, which were similar to conditions during infection, expressed 68 genes associated primarily with innate immune activation and neutrophil recruitment. In contrast, FLS stimulated with IFNγ, which were similar to conditions in the postinfectious phase, expressed >2,000 genes associated with pathogen sensing, inflammation, and MHC Class II antigen presentation, similar to the expression profile in postinfectious synovial tissue. Furthermore, costimulation of FLS with B. burgdorferi and IFNγ induced greater expression of IL-6 and other innate immune response proteins and genes than with IFNγ stimulation alone. These results suggest that B. burgdorferi infection, in combination with IFNγ, initiates the differentiation of FLS into a highly inflammatory phenotype. We hypothesise that overexpression of IFNγ by lymphocytes within synovia perpetuates these responses in the postinfectious period, causing proliferative synovitis and stalling appropriate repair of damaged tissue.
Collapse
Affiliation(s)
- Robert B Lochhead
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - David Ordoñez
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sheila L Arvikar
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - John M Aversa
- Department of Orthopedics, Yale University School of Medicine, New Haven, Connecticut
| | - Luke S Oh
- Department of Orthopedics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Benton Heyworth
- Department of Orthopedics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ruslan Sadreyev
- Department of Molecular Biology and Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Klemen Strle
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Dubrot J, Duraes FV, Harlé G, Schlaeppi A, Brighouse D, Madelon N, Göpfert C, Stokar-Regenscheit N, Acha-Orbea H, Reith W, Gannagé M, Hugues S. Absence of MHC-II expression by lymph node stromal cells results in autoimmunity. Life Sci Alliance 2018; 1:e201800164. [PMID: 30584641 PMCID: PMC6297861 DOI: 10.26508/lsa.201800164] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 12/18/2022] Open
Abstract
MHCII-restricted antigen presentation by lymph node stromal cells is essential for regulatory T-cell proliferation and functions, and for the regulation of autoimmunity. How lymph node stromal cells (LNSCs) shape peripheral T-cell responses remains unclear. We have previously demonstrated that murine LNSCs, lymphatic endothelial cells (LECs), blood endothelial cells (BECs), and fibroblastic reticular cells (FRCs) use the IFN-γ–inducible promoter IV (pIV) of the MHC class II (MHCII) transactivator CIITA to express MHCII. Here, we show that aging mice (>1 yr old) in which MHCII is abrogated in LNSCs by the selective deletion of pIV exhibit a significant T-cell dysregulation in LNs, including defective Treg and increased effector CD4+ and CD8+ T-cell frequencies, resulting in enhanced peripheral organ T-cell infiltration and autoantibody production. The proliferation of LN-Tregs interacting with LECs increases following MHCII up-regulation by LECs upon aging or after exposure to IFN-γ, this effect being abolished in mice in which LECs lack MHCII. Overall, our work underpins the importance of LNSCs, particularly LECs, in supporting Tregs and T-cell tolerance.
Collapse
Affiliation(s)
- Juan Dubrot
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Fernanda V Duraes
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Guillaume Harlé
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Anjalie Schlaeppi
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Natacha Madelon
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland.,Division of Rheumatology, Department of Internal Medicine, University Hospital Geneva, Geneva, Switzerland
| | - Christine Göpfert
- Institute of Animal Pathology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nadine Stokar-Regenscheit
- Institute of Animal Pathology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Hans Acha-Orbea
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Walter Reith
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Monique Gannagé
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland.,Division of Rheumatology, Department of Internal Medicine, University Hospital Geneva, Geneva, Switzerland
| | - Stephanie Hugues
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
22
|
Williams GP, Schonhoff AM, Jurkuvenaite A, Thome AD, Standaert DG, Harms AS. Targeting of the class II transactivator attenuates inflammation and neurodegeneration in an alpha-synuclein model of Parkinson's disease. J Neuroinflammation 2018; 15:244. [PMID: 30165873 PMCID: PMC6117927 DOI: 10.1186/s12974-018-1286-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/20/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by intracellular alpha-synuclein (α-syn) inclusions, progressive death of dopaminergic neurons in the substantia nigra pars compacta (SNpc), and activation of the innate and adaptive immune systems. Disruption of immune signaling between the central nervous system (CNS) and periphery, such as through targeting the chemokine receptor type 2 (CCR2) or the major histocompatibility complex II (MHCII), is neuroprotective in rodent models of PD, suggesting a key role for innate and adaptive immunity in disease progression. The purpose of this study was to investigate whether genetic knockout or RNA silencing of the class II transactivator (CIITA), a transcriptional co-activator required for MHCII induction, is effective in reducing the neuroinflammation and neurodegeneration observed in an α-syn mouse model of PD. METHODS In vitro, we utilized microglia cultures from WT or CIITA -/- mice treated with α-syn fibrils to investigate inflammatory iNOS expression and antigen processing via immunocytochemistry (ICC). In vivo, an adeno-associated virus (AAV) was used to overexpress α-syn in WT and CIITA -/- mice as a model for PD. Concurrently with AAV-mediated overexpression of α-syn, WT mice received CIITA-targeted shRNAs packaged in lentiviral constructs. Immunohistochemistry and flow cytometry were used to assess inflammation and peripheral cell infiltration at 4 weeks post transduction, and unbiased stereology was used 6 months post transduction to assess neurodegeneration. RESULTS Using ICC and DQ-ovalbumin, we show that CIITA -/- microglial cultures failed to upregulate iNOS and MHCII expression, and had decreased antigen processing in response to α-syn fibrils when compared to WT microglia. In vivo, global knock-out of CIITA as well as local knockdown using lentiviral shRNAs targeting CIITA attenuated MHCII expression, peripheral immune cell infiltration, and α-syn-induced neurodegeneration. CONCLUSION Our data provide evidence that CIITA is required for α-syn-induced MHCII induction and subsequent infiltration of peripheral immune cells in an α-syn mouse model of PD. Additionally, we demonstrate that CIITA in the CNS drives neuroinflammation and neurodegeneration. These data provide further support that the disruption or modulation of antigen processing and presentation via CIITA is a promising target for therapeutic development in preclinical animal models of PD.
Collapse
Affiliation(s)
- Gregory P Williams
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham (UAB), 1719 6th Ave. South, CIRC 525, Birmingham, AL, 35294-0021, USA
| | - Aubrey M Schonhoff
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham (UAB), 1719 6th Ave. South, CIRC 525, Birmingham, AL, 35294-0021, USA
| | - Asta Jurkuvenaite
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham (UAB), 1719 6th Ave. South, CIRC 525, Birmingham, AL, 35294-0021, USA
| | - Aaron D Thome
- Department of Neurology, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - David G Standaert
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham (UAB), 1719 6th Ave. South, CIRC 525, Birmingham, AL, 35294-0021, USA
| | - Ashley S Harms
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham (UAB), 1719 6th Ave. South, CIRC 525, Birmingham, AL, 35294-0021, USA.
| |
Collapse
|
23
|
Berglund AK, Fisher MB, Cameron KA, Poole EJ, Schnabel LV. Transforming Growth Factor-β2 Downregulates Major Histocompatibility Complex (MHC) I and MHC II Surface Expression on Equine Bone Marrow-Derived Mesenchymal Stem Cells Without Altering Other Phenotypic Cell Surface Markers. Front Vet Sci 2017; 4:84. [PMID: 28660198 PMCID: PMC5466990 DOI: 10.3389/fvets.2017.00084] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022] Open
Abstract
Allogeneic mesenchymal stem cells (MSCs) are a promising cell source for treating musculoskeletal injuries in horses. Effective and safe allogeneic therapy may be hindered, however, by recipient immune recognition and rejection of major histocompatibility complex (MHC)-mismatched MSCs. Development of strategies to prevent immune rejection of MHC-mismatched MSCs in vivo is necessary to enhance cell survival and potentially increase the efficacy and safety of allogeneic MSC therapy. The purposes of this study were to evaluate if transforming growth factor-β2 (TGF-β2) downregulated MHC expression on equine MSCs and to determine if TGF-β2 treatment altered the phenotype of MSCs. Equine bone marrow-derived MSCs from 12 horses were treated with 1, 5, or 10 ng/ml TGF-β2 from initial isolation until MHC expression analysis. TGF-β2-treated MSCs had reduced MHC I and MHC II surface expression compared to untreated controls. TGF-β2 treatment also partially blocked IFN-γ-induced upregulation of MHC I and MHC II. Constitutive and IFN-γ-induced MHC I and MHC II expression on equine MSCs was dynamic and highly variable, and the effect of TGF-β2 was significantly dependent on the donor animal and baseline MHC expression. TGF-β2 treatment did not appear to change morphology, surface marker expression, MSC viability, or secretion of TGF-β1, but did significantly increase the number of cells obtained from culture. These results indicate that TGF-β2 treatment has promise for regulating MHC expression on MSCs to facilitate allogeneic therapy, but further work is needed to maintain MHC stability when exposed to an inflammatory stimulus.
Collapse
Affiliation(s)
- Alix K. Berglund
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Matthew B. Fisher
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Department of Biomedical Engineering, North Carolina State University, Raleigh, and University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristin A. Cameron
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Emma J. Poole
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Lauren V. Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
24
|
Chelbi S, Dang A, Guarda G. Emerging Major Histocompatibility Complex Class I-Related Functions of NLRC5. Adv Immunol 2017; 133:89-119. [DOI: 10.1016/bs.ai.2016.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
25
|
Robinette ML, Colonna M. Innate lymphoid cells and the MHC. HLA 2016; 87:5-11. [PMID: 26812060 DOI: 10.1111/tan.12723] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 11/24/2015] [Indexed: 12/29/2022]
Abstract
Innate lymphoid cells (ILCs) are a new class of immune cells that include natural killer (NK) cells and appear to be the innate counterparts to CD4(+) helper T cells and CD8(+) cytotoxic T cells based on developmental and functional similarities. Like T cells, both NK cells and other ILCs also show connections to the major histocompatibility complex (MHC). In human and mouse, NK cells recognize and respond to classical and nonclassical MHC I molecules as well as structural homologues, whereas mouse ILCs have recently been shown to express MHC II. We describe the history of MHC I recognition by NK cells and discuss emerging roles for MHC II expression by ILC subsets, making comparisons between both mouse and human when possible.
Collapse
Affiliation(s)
- M L Robinette
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - M Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
26
|
Puvvada S, Li H, Rimsza LM, Bernstein SH, Fisher RI, LeBlanc M, Schmelz M, Glinsmann-Gibson B, Miller TP, Maddox AM, Friedberg JW, Smith SM, Persky DO. A phase II study of belinostat (PXD101) in relapsed and refractory aggressive B-cell lymphomas: SWOG S0520. Leuk Lymphoma 2016; 57:2359-69. [PMID: 26758422 PMCID: PMC5140034 DOI: 10.3109/10428194.2015.1135431] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recent advances in diffuse large B-cell lymphomas (DLBCL) have underscored the importance of tumor microenvironment in escaping host anti-tumor responses. One mechanism is loss of major histocompatibility Class II antigens (MHCII) associated with decreased tumor infiltrating T lymphocytes (TIL) and poor survival. Transcription of MHCII is controlled by CIITA which in turn is regulated by histone acetylation. In this study, we hypothesized that HDAC inhibition with belinostat increases MHCII, CIITA expression, TIL and improves patient outcomes. Primary objective was evaluation of toxicity and response. Twenty-two patients were enrolled for the study. Belinostat was well tolerated with mild toxicity. Two partial responses were observed at 5, 13 months after registration for an overall response rate (ORR) (95% CI) of 10.5% (1.3-33.1%), and three patients had stable disease for 4.7, 42.3+, and 68.4 + months with minimum 3-year follow-up. Included correlative studies support the hypothesis and serve as the basis for SWOG S0806 combining vorinostat with R-CHOP.
Collapse
Affiliation(s)
| | - Hongli Li
- SWOG Statistical Center, Seattle, WA
| | - Lisa M. Rimsza
- Department of Pathology, University of Arizona, Tucson, AZ
| | | | | | | | - Monika Schmelz
- Department of Pathology, University of Arizona, Tucson, AZ
| | | | | | | | | | | | | |
Collapse
|
27
|
Major Histocompatibility Complex Class II HLA-DRα Is Downregulated by Kaposi's Sarcoma-Associated Herpesvirus-Encoded Lytic Transactivator RTA and MARCH8. J Virol 2016; 90:8047-58. [PMID: 27356905 DOI: 10.1128/jvi.01079-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 06/07/2016] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Kaposi's sarcoma-associated herpesvirus (KSHV) maintains two modes of life cycle, the latent and lytic phases. To evade the attack of the cell host's immune system, KSHV switches from the lytic to the latent phase, a phase in which only a few of viral proteins are expressed. The mechanism by which KSHV evades the attack of the immune system and establishes latency has not been fully understood. Major histocompatibility complex class II (MHC-II) molecules are key components of the immune system defense mechanism against viral infections. Here we report that HLA-DRα, a member of the MHC-II molecules, was downregulated by the replication and transcription activator (RTA) protein encoded by KSHV ORF50, an important regulator of the viral life cycle. RTA not only downregulated HLA-DRα at the protein level through direct binding and degradation through the proteasome pathway but also indirectly downregulated the protein level of HLA-DRα by enhancing the expression of MARCH8, a member of the membrane-associated RING-CH (MARCH) proteins. Our findings indicate that KSHV RTA facilitates evasion of the virus from the immune system through manipulation of HLA-DRα. IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) has a causal role in a number of human cancers, and its persistence in infected cells is controlled by the host's immune system. The mechanism by which KSHV evades an attack by the immune system has not been well understood. This work represents studies which identify a novel mechanism by which the virus can facilitate evasion of an immune system. We now show that RTA, the replication and transcription activator encoded by KSHV (ORF50), can function as an E3 ligase to degrade HLA-DRα. It can directly bind and induce degradation of HLA-DRα through the ubiquitin-proteasome degradation pathway. In addition to the direct regulation of HLA-DRα, RTA can also indirectly downregulate the level of HLA-DRα protein by upregulating transcription of MARCH8. Increased MARCH8 results in the downregulation of HLA-DRα. Furthermore, we also demonstrate that expression of HLA-DRα was impaired in KSHV de novo infection.
Collapse
|
28
|
Thelemann C, Haller S, Blyszczuk P, Kania G, Rosa M, Eriksson U, Rotman S, Reith W, Acha-Orbea H. Absence of nonhematopoietic MHC class II expression protects mice from experimental autoimmune myocarditis. Eur J Immunol 2015; 46:656-64. [PMID: 26621778 DOI: 10.1002/eji.201545945] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/26/2015] [Accepted: 11/25/2015] [Indexed: 01/14/2023]
Abstract
Experimental autoimmune myocarditis (EAM) is a CD4(+) T-cell-mediated model of human inflammatory dilated cardiomyopathies. Heart-specific CD4(+) T-cell activation is dependent on autoantigens presented by MHC class II (MHCII) molecules expressed on professional APCs. In this study, we addressed the role of inflammation-induced MHCII expression by cardiac nonhematopoietic cells on EAM development. EAM was induced in susceptible mice lacking inducible expression of MHCII molecules on all nonhematopoietic cells (pIV-/- K14 class II transactivator (CIITA) transgenic (Tg) mice) by immunization with α-myosin heavy chain peptide in CFA. Lack of inducible nonhematopoietic MHCII expression in pIV-/- K14 CIITA Tg mice conferred EAM resistance. In contrast, cardiac pathology was induced in WT and heterozygous mice, and correlated with elevated cardiac endothelial MHCII expression. Control mice with myocarditis displayed an increase in infiltrating CD4(+) T cells and in expression of IFN-γ, which is the major driver of nonhematopoietic MHCII expression. Mechanistically, IFN-γ neutralization in WT mice shortly before disease onset resulted in reduced cardiac MHCII expression and pathology. These findings reveal a previously overlooked contribution of IFN-γ to induce endothelial MHCII expression in the heart and to progress cardiac pathology during myocarditis.
Collapse
Affiliation(s)
- Christoph Thelemann
- Department of Biochemistry, CIIL, University of Lausanne, Epalinges, Switzerland
| | - Sergio Haller
- Department of Biochemistry, CIIL, University of Lausanne, Epalinges, Switzerland
| | - Przemyslaw Blyszczuk
- Division of Cardioimmunology, Centre of Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Gabriela Kania
- Research of Systemic Autoimmune Diseases, Division of Rheumatology, University Hospital Zürich, Schlieren, Switzerland
| | - Muriel Rosa
- Department of Biochemistry, CIIL, University of Lausanne, Epalinges, Switzerland
| | - Urs Eriksson
- Division of Cardioimmunology, Centre of Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Samuel Rotman
- Institute of Pathology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - Walter Reith
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Hans Acha-Orbea
- Department of Biochemistry, CIIL, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
29
|
Lee A, Park SP, Park CH, Kang BH, Park SH, Ha SJ, Jung KC. IL-4 Induced Innate CD8+ T Cells Control Persistent Viral Infection. PLoS Pathog 2015; 11:e1005193. [PMID: 26452143 PMCID: PMC4599894 DOI: 10.1371/journal.ppat.1005193] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 09/06/2015] [Indexed: 12/20/2022] Open
Abstract
Memory-like CD8+ T cells expressing eomesodermin are a subset of innate T cells initially identified in a number of genetically modified mice, and also exist in wild mice and human. The acquisition of memory phenotype and function by these T cells is dependent on IL–4 produced by PLZF+ innate T cells; however, their physiologic function is still not known. Here we found that these IL-4-induced innate CD8+ T cells are critical for accelerating the control of chronic virus infection. In CIITA-transgenic mice, which have a substantial population of IL-4-induced innate CD8+ T cells, this population facilitated rapid control of viremia and induction of functional anti-viral T-cell responses during infection with chronic form of lymphocytic choriomeningitis virus. Characteristically, anti-viral innate CD8+ T cells accumulated sufficiently during early phase of infection. They produced a robust amount of IFN-γ and TNF-α with enhanced expression of a degranulation marker. Furthermore, this finding was confirmed in wild-type mice. Taken together, the results from our study show that innate CD8+ T cells works as an early defense mechanism against chronic viral infection. Over the course of viral infection there may be a limited time period during which the host system can eliminate the virus. When viruses are not eliminated within this period of time, virus can establish persistent infection. Here, we show that IL-4-induced innate CD8+ T cells are able to effectively control chronic virus infection. Innate T cells are heterogeneous population of T cells that acquire effector/memory phenotype as a result of their maturation process in thymus, unlike conventional T cells that differentiate into memory cells after antigen encounter in periphery. Previous data suggest that innate T cells might serve as a first-line of defense against certain bacterial pathogens. IL-4-induced innate CD8+ T cells are a unique subset of innate T cells that were recently identified in both mouse and human. We found that IL-4-induced innate CD8+ T cells immediately accumulated after viral infection and produced a robust amount of effector cytokines. Thereby, IL-4-induced innate CD8+ T cells provide an effective barrier to the establishment of persistent infection via effective virus control during the early phase of viral infection. Collectively our data show that IL-4-induced innate CD8+ T cells works as an early defense mechanism against chronic viral infection.
Collapse
Affiliation(s)
- Ara Lee
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
| | - Seung Pyo Park
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Chan Hee Park
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
| | - Byung Hyun Kang
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seong Hoe Park
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
- * E-mail: (SJH); (KCJ)
| | - Kyeong Cheon Jung
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- * E-mail: (SJH); (KCJ)
| |
Collapse
|
30
|
Hirosue S, Dubrot J. Modes of Antigen Presentation by Lymph Node Stromal Cells and Their Immunological Implications. Front Immunol 2015; 6:446. [PMID: 26441957 PMCID: PMC4561840 DOI: 10.3389/fimmu.2015.00446] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 08/17/2015] [Indexed: 12/15/2022] Open
Abstract
Antigen presentation is no longer the exclusive domain of cells of hematopoietic origin. Recent works have demonstrated that lymph node stromal cell (LNSC) populations, such as fibroblastic reticular cells, lymphatic and blood endothelial cells, not only provide a scaffold for lymphocyte interactions but also exhibit active immunomodulatory roles that are critical to mounting and resolving effective immune responses. Importantly, LNSCs possess the ability to present antigens and establish antigen-specific interactions with T cells. One example is the expression of peripheral tissue antigens, which are presented on major histocompatibility complex (MHC)-I molecules with tolerogenic consequences on T cells. Additionally, exogenous antigens, including self and tumor antigens, can be processed and presented on MHC-I complexes, which result in dysfunctional activation of antigen-specific CD8+ T cells. While MHC-I is widely expressed on cells of both hematopoietic and non-hematopoietic origins, antigen presentation via MHC-II is more precisely regulated. Nevertheless, LNSCs are capable of endogenously expressing, or alternatively, acquiring MHC-II molecules. Transfer of antigen between LNSC and dendritic cells in both directions has been recently suggested to promote tolerogenic roles of LNSCs on the CD4+ T cell compartment. Thus, antigen presentation by LNSCs is thought to be a mechanism that promotes the maintenance of peripheral tolerance as well as generates a pool of diverse antigen-experienced T cells for protective immunity. This review aims to integrate the current and emerging literature to highlight the importance of LNSCs in immune responses, and emphasize their role in antigen trafficking, retention, and presentation.
Collapse
Affiliation(s)
- Sachiko Hirosue
- Institute of Bioengineering, École Polytechnique Fédéral de Lausanne , Lausanne , Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, Université de Genève , Geneva , Switzerland
| |
Collapse
|
31
|
Jun H, Ying H, Daiwen C, Bing Y, Xiangbing M, Ping Z, Jie Y, Zhiqing H, Junqiu L. miR-628, a microRNA that is induced by Toll-like receptor stimulation, regulates porcine innate immune responses. Sci Rep 2015; 5:12226. [PMID: 26227240 PMCID: PMC4521185 DOI: 10.1038/srep12226] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/22/2015] [Indexed: 12/24/2022] Open
Abstract
Mammalian innate and acquired immune responses involve a coordinated, sequential, and self limiting sequence of events controlled by positive and negative regulatory mechanism. MicroRNAs have been implicated as a negative regulator for diverse biological events including immune responses. However, the involvement of miRNAs in regulating the immune responses is just beginning to be explored. Here, we characterized the expression profiling of 375 microRNAs in porcine monocytes induced by lipopolysaccharide (LPS), and result shows that several of them are endotoxin-responsive genes. Through promoter analysis, the miR-628 was found to be a NF-κB dependent gene. Importantly, miR-628 was predicted to base-pair with sequences in the 3'-UTR of the myeloid differentiation protein 88 (MyD88) gene. And we found that the UTR inhibit expression of a linked reporter gene coding a key adapter molecule downstream of Toll-like receptors (TLRs), resulting in suppressing of the TLR signaling. Therefore, we not only propose a role of miR-628 in control of the TLR signaling through a negative feedback regulation loop involving down-regulation of MyD88 protein levels, but results may also contribute to rational target selection orchestrating the inflammatory responses.
Collapse
Affiliation(s)
- He Jun
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - He Ying
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Chen Daiwen
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Yu Bing
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Mao Xiangbing
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Zheng Ping
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Yu Jie
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Huang Zhiqing
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| | - Luo Junqiu
- 1] Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan 625014, P. R. China [2] Key Laborotary of Animal Disease-Resistance Nutrition, Ministry of Education, China
| |
Collapse
|
32
|
Hepworth MR, Fung TC, Masur SH, Kelsen JR, McConnell FM, Dubrot J, Withers DR, Hugues S, Farrar MA, Reith W, Eberl G, Baldassano RN, Laufer TM, Elson CO, Sonnenberg GF. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4⁺ T cells. Science 2015; 348:1031-5. [PMID: 25908663 PMCID: PMC4449822 DOI: 10.1126/science.aaa4812] [Citation(s) in RCA: 391] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/07/2015] [Indexed: 12/12/2022]
Abstract
Inflammatory CD4(+) T cell responses to self or commensal bacteria underlie the pathogenesis of autoimmunity and inflammatory bowel disease (IBD), respectively. Although selection of self-specific T cells in the thymus limits responses to mammalian tissue antigens, the mechanisms that control selection of commensal bacteria-specific T cells remain poorly understood. Here, we demonstrate that group 3 innate lymphoid cell (ILC3)-intrinsic expression of major histocompatibility complex class II (MHCII) is regulated similarly to thymic epithelial cells and that MHCII(+) ILC3s directly induce cell death of activated commensal bacteria-specific T cells. Further, MHCII on colonic ILC3s was reduced in pediatric IBD patients. Collectively, these results define a selection pathway for commensal bacteria-specific CD4(+) T cells in the intestine and suggest that this process is dysregulated in human IBD.
Collapse
Affiliation(s)
- Matthew R Hepworth
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Gastroenterology Division, and Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Thomas C Fung
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Gastroenterology Division, and Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, USA. Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel H Masur
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Judith R Kelsen
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fiona M McConnell
- Medical Research Council, Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - David R Withers
- Medical Research Council, Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Stephanie Hugues
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Michael A Farrar
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, MN, USA
| | - Walter Reith
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Gérard Eberl
- Institut Pasteur, Microenvironment and Immunity Unit, Paris, France
| | - Robert N Baldassano
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Terri M Laufer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Charles O Elson
- Departments of Medicine and Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Gastroenterology Division, and Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| |
Collapse
|
33
|
Inhibins tune the thymocyte selection process by regulating thymic stromal cell differentiation. J Immunol Res 2015; 2015:837859. [PMID: 25973437 PMCID: PMC4418002 DOI: 10.1155/2015/837859] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 01/15/2015] [Accepted: 01/20/2015] [Indexed: 01/01/2023] Open
Abstract
Inhibins and Activins are members of the TGF-β superfamily that regulate the differentiation of several cell types. These ligands were initially identified as hormones that regulate the hypothalamus-pituitary-gonadal axis; however, increasing evidence has demonstrated that they are key regulators in the immune system. We have previously demonstrated that Inhibins are the main Activin ligands expressed in the murine thymus and that they regulate thymocyte differentiation, promoting the DN3-DN4 transition and the selection of SP thymocytes. As Inhibins are mainly produced by thymic stromal cells, which also express Activin receptors and Smad proteins, we hypothesized that Inhibins might play a role in stromal cell differentiation and function. Here, we demonstrate that, in the absence of Inhibins, thymic conventional dendritic cells display reduced levels of MHC Class II (MHCII) and CD86. In addition, the ratio between cTECs and mTECs was affected, indicating that mTEC differentiation was favoured and cTEC diminished in the absence of Inhibins. These changes appeared to impact thymocyte selection leading to a decreased selection of CD4SP thymocytes and increased generation of natural regulatory T cells. These findings demonstrate that Inhibins tune the T cell selection process by regulating both thymocyte and stromal cell differentiation.
Collapse
|
34
|
Guéry L, Dubrot J, Lippens C, Brighouse D, Malinge P, Irla M, Pot C, Reith W, Waldburger JM, Hugues S. Ag-presenting CpG-activated pDCs prime Th17 cells that induce tumor regression. Cancer Res 2014; 74:6430-40. [PMID: 25252912 DOI: 10.1158/0008-5472.can-14-1149] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Plasmacytoid dendritic cells (pDC) rapidly and massively produce type I IFN and other inflammatory cytokines in response to foreign nucleic acids, thereby indirectly influencing T-cell responses. Moreover, antigen (Ag)-presenting pDCs directly regulate T-cell differentiation. Depending on the immune environment, pDCs exhibit either tolerogenic or immunogenic properties. Here, we show that CpG-activated pDCs promote efficient Th17 differentiation. Indeed, Th17 responses are defective in mice selectively lacking MHCII on pDCs upon antigenic challenge. Importantly, in those mice, the frequency of Th17 cells infiltrating solid tumors is impaired. As a result, the recruitment of infiltrating leukocytes in tumors, including tumor-specific cytotoxic T lymphocytes (CTL), is altered and results in increased tumor growth. Importantly, following immunization with tumor Ag and CpG-B, MHCII-restricted Ag presentation by pDCs promotes the differentiation of antitumor Th17 cells that induce intratumor CTL recruitment and subsequent regression of established tumors. Our results highlight a new role for Ag presenting activated pDCs in promoting the development of Th17 cells and impacting on antitumor immunity.
Collapse
Affiliation(s)
- Leslie Guéry
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Carla Lippens
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | | | - Magali Irla
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland. Centre d'immunology de Marseille Luminy, Université de la Méditerranée, Marseille, France
| | - Caroline Pot
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland. Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Walter Reith
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Jean-Marc Waldburger
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland.
| |
Collapse
|
35
|
Benasciutti E, Mariani E, Oliva L, Scolari M, Perilli E, Barras E, Milan E, Orfanelli U, Fazzalari NL, Campana L, Capobianco A, Otten L, Particelli F, Acha-Orbea H, Baruffaldi F, Faccio R, Sitia R, Reith W, Cenci S. MHC class II transactivator is an in vivo regulator of osteoclast differentiation and bone homeostasis co-opted from adaptive immunity. J Bone Miner Res 2014; 29:290-303. [PMID: 24038328 DOI: 10.1002/jbmr.2090] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 07/30/2013] [Accepted: 08/22/2013] [Indexed: 12/26/2022]
Abstract
The molecular networks controlling bone homeostasis are not fully understood. The common evolution of bone and adaptive immunity encourages the investigation of shared regulatory circuits. MHC Class II Transactivator (CIITA) is a master transcriptional co-activator believed to be exclusively dedicated for antigen presentation. CIITA is expressed in osteoclast precursors, and its expression is accentuated in osteoporotic mice. We thus asked whether CIITA plays a role in bone biology. To this aim, we fully characterized the bone phenotype of two mouse models of CIITA overexpression, respectively systemic and restricted to the monocyte-osteoclast lineage. Both CIITA-overexpressing mouse models revealed severe spontaneous osteoporosis, as assessed by micro-computed tomography and histomorphometry, associated with increased osteoclast numbers and enhanced in vivo bone resorption, whereas osteoblast numbers and in vivo bone-forming activity were unaffected. To understand the underlying cellular and molecular bases, we investigated ex vivo the differentiation of mutant bone marrow monocytes into osteoclasts and immune effectors, as well as osteoclastogenic signaling pathways. CIITA-overexpressing monocytes differentiated normally into effector macrophages or dendritic cells but showed enhanced osteoclastogenesis, whereas CIITA ablation suppressed osteoclast differentiation. Increased c-fms and receptor activator of NF-κB (RANK) signaling underlay enhanced osteoclast differentiation from CIITA-overexpressing precursors. Moreover, by extending selected phenotypic and cellular analyses to additional genetic mouse models, namely MHC Class II deficient mice and a transgenic mouse line lacking a specific CIITA promoter and re-expressing CIITA in the thymus, we excluded MHC Class II expression and T cells from contributing to the observed skeletal phenotype. Altogether, our study provides compelling genetic evidence that CIITA, the molecular switch of antigen presentation, plays a novel, unexpected function in skeletal homeostasis, independent of MHC Class II expression and T cells, by exerting a selective and intrinsic control of osteoclast differentiation and bone resorption in vivo.
Collapse
Affiliation(s)
- Elisa Benasciutti
- Division of Genetics and Cell Biology and BoNetwork, DiBiT, San Raffaele Scientific Institute, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Interferon-γ induces expression of MHC class II on intestinal epithelial cells and protects mice from colitis. PLoS One 2014; 9:e86844. [PMID: 24489792 PMCID: PMC3904943 DOI: 10.1371/journal.pone.0086844] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/13/2013] [Indexed: 12/12/2022] Open
Abstract
Immune responses against intestinal microbiota contribute to the pathogenesis of inflammatory bowel diseases (IBD) and involve CD4+ T cells, which are activated by major histocompatibility complex class II (MHCII) molecules on antigen-presenting cells (APCs). However, it is largely unexplored how inflammation-induced MHCII expression by intestinal epithelial cells (IEC) affects CD4+ T cell-mediated immunity or tolerance induction in vivo. Here, we investigated how epithelial MHCII expression is induced and how a deficiency in inducible epithelial MHCII expression alters susceptibility to colitis and the outcome of colon-specific immune responses. Colitis was induced in mice that lacked inducible expression of MHCII molecules on all nonhematopoietic cells, or specifically on IECs, by continuous infection with Helicobacter hepaticus and administration of interleukin (IL)-10 receptor-blocking antibodies (anti-IL10R mAb). To assess the role of interferon (IFN)-γ in inducing epithelial MHCII expression, the T cell adoptive transfer model of colitis was used. Abrogation of MHCII expression by nonhematopoietic cells or IECs induces colitis associated with increased colonic frequencies of innate immune cells and expression of proinflammatory cytokines. CD4+ T-helper type (Th)1 cells - but not group 3 innate lymphoid cells (ILCs) or Th17 cells - are elevated, resulting in an unfavourably altered ratio between CD4+ T cells and forkhead box P3 (FoxP3)+ regulatory T (Treg) cells. IFN-γ produced mainly by CD4+ T cells is required to upregulate MHCII expression by IECs. These results suggest that, in addition to its proinflammatory roles, IFN-γ exerts a critical anti-inflammatory function in the intestine which protects against colitis by inducing MHCII expression on IECs. This may explain the failure of anti-IFN-γ treatment to induce remission in IBD patients, despite the association of elevated IFN-γ and IBD.
Collapse
|
37
|
Duraes FV, Thelemann C, Sarter K, Acha-Orbea H, Hugues S, Reith W. Role of major histocompatibility complex class II expression by non-hematopoietic cells in autoimmune and inflammatory disorders: facts and fiction. ACTA ACUST UNITED AC 2014; 82:1-15. [PMID: 23745569 DOI: 10.1111/tan.12136] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It is well established that interactions between CD4(+) T cells and major histocompatibility complex class II (MHCII) positive antigen-presenting cells (APCs) of hematopoietic origin play key roles in both the maintenance of tolerance and the initiation and development of autoimmune and inflammatory disorders. In sharp contrast, despite nearly three decades of intensive research, the functional relevance of MHCII expression by non-hematopoietic tissue-resident cells has remained obscure. The widespread assumption that MHCII expression by non-hematopoietic APCs has an impact on autoimmune and inflammatory diseases has in most instances neither been confirmed nor excluded by indisputable in vivo data. Here we review and put into perspective conflicting in vitro and in vivo results on the putative impact of MHCII expression by non-hematopoietic APCs--in both target organs and secondary lymphoid tissues--on the initiation and development of representative autoimmune and inflammatory disorders. Emphasis will be placed on the lacunar status of our knowledge in this field. We also discuss new mouse models--developed on the basis of our understanding of the molecular mechanisms that regulate MHCII expression--that constitute valuable tools for filling the severe gaps in our knowledge on the functions of non-hematopoietic APCs in inflammatory conditions.
Collapse
Affiliation(s)
- F V Duraes
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
38
|
Cai Q, Banerjee S, Cervini A, Lu J, Hislop AD, Dzeng R, Robertson ES. IRF-4-mediated CIITA transcription is blocked by KSHV encoded LANA to inhibit MHC II presentation. PLoS Pathog 2013; 9:e1003751. [PMID: 24204280 PMCID: PMC3814934 DOI: 10.1371/journal.ppat.1003751] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/23/2013] [Indexed: 01/18/2023] Open
Abstract
Peptides presentation to T cells by MHC class II molecules is of importance in initiation of immune response to a pathogen. The level of MHC II expression directly influences T lymphocyte activation and is often targeted by various viruses. Kaposi's sarcoma-associated herpesvirus (KSHV) encoded LANA is known to evade MHC class I peptide processing, however, the effect of LANA on MHC class II remains unclear. Here, we report that LANA down-regulates MHC II expression and presentation by inhibiting the transcription of MHC II transactivator (CIITA) promoter pIII and pIV in a dose-dependent manner. Strikingly, although LANA knockdown efficiently disrupts the inhibition of CIITA transcripts from its pIII and pIV promoter region, the expression of HLA-DQβ but no other MHC II molecules was significantly restored. Moreover, we revealed that the presentation of HLA-DQβ enhanced by LANA knockdown did not help LANA-specific CD4+ T cell recognition of PEL cells, and the inhibition of CIITA by LANA is independent of IL-4 or IFN-γ signaling but dependent on the direct interaction of LANA with IRF-4 (an activator of both the pIII and pIV CIITA promoters). This interaction dramatically blocked the DNA-binding ability of IRF-4 on both pIII and pIV promoters. Thus, our data implies that LANA can evade MHC II presentation and suppress CIITA transcription to provide a unique strategy of KSHV escape from immune surveillance by cytotoxic T cells.
Collapse
Affiliation(s)
- Qiliang Cai
- MOE&MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Shuvomoy Banerjee
- Department of Microbiology and the Tumor Virology Program of Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States of America
| | - Amanda Cervini
- Department of Microbiology and the Tumor Virology Program of Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States of America
| | - Jie Lu
- Department of Microbiology and the Tumor Virology Program of Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States of America
| | - Andrew D. Hislop
- School of Cancer Sciences and Medical Research Council Centre for Immune Regulation, The University of Birmingham, Birmingham, United Kingdom
| | - Richard Dzeng
- Department of Microbiology and the Tumor Virology Program of Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States of America
| | - Erle S. Robertson
- Department of Microbiology and the Tumor Virology Program of Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States of America
| |
Collapse
|
39
|
Fang M, Xia J, Wu X, Kong H, Wang H, Xie W, Xu Y. Adenosine signaling inhibits CIITA-mediated MHC class II transactivation in lung fibroblast cells. Eur J Immunol 2013; 43:2162-73. [PMID: 23681904 DOI: 10.1002/eji.201343461] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/09/2013] [Accepted: 05/13/2013] [Indexed: 01/28/2023]
Abstract
Efficient antigen presentation by major histocompatibility complex (MHC) molecules represents a critical process in adaptive immunity. Class II transactivator (CIITA) is considered the master regulator of MHC class II (MHC II) transcription. Previously, we have shown that CIITA expression is upregulated in smooth muscle cells deficient in A2b adenosine receptor. Here, we report that treatment with the adenosine receptor agonist adenosine-5'N-ethylcarboxamide (NECA) attenuated MHC II transcription in lung fibro-blast cells as a result of CIITA repression. Further analysis revealed that NECA preferentially abrogated CIITA transcription through promoters III and IV. Blockade with a selective A2b receptor antagonist MRS-1754 restored CIITA-dependent MHC II transactivation. Forskolin, an adenylyl cyclase activator, achieved the same effect as NECA. A2b signaling repressed CIITA transcription by altering histone modifications and recruitment of key factors on the CIITA promoters in a STAT1-dependent manner. MRS-1754 blocked the antagonism of transforming growth factor beta (TGF-β) in CIITA induction by interferon gamma (IFN-γ), alluding to a potential dialogue between TGF-β and adenosine signaling pathways. Finally, A2b signaling attenuated STAT1 phosphorylation and stimulated TGF-β synthesis. In conclusion, we have identified an adenosine-A2b receptor-adenylyl cyclase axis that influences CIITA-mediated MHC II transactivation in lung fibroblast cells and as such have provided invaluable insights into the development of novel immune-modulatory strategies.
Collapse
Affiliation(s)
- Mingming Fang
- Department of Surgery, Jiangsu Jiankang Vocational College, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Waldburger JM, Palmer G, Seemayer C, Lamacchia C, Finckh A, Christofilopoulos P, Baeten D, Reith W, Gabay C. Autoimmunity and inflammation are independent of class II transactivator type PIV-dependent class II major histocompatibility complex expression in peripheral tissues during collagen-induced arthritis. ACTA ACUST UNITED AC 2013; 63:3354-63. [PMID: 21739421 DOI: 10.1002/art.30522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To determine the regulation of class II major histocompatibility complex (MHC) expression in fibroblast-like synoviocytes (FLS) in order to investigate their role as nonprofessional antigen-presenting cells in collagen-induced arthritis (CIA). METHODS Expression of class II MHC, class II MHC transactivator (CIITA), and Ciita isoforms PI, PIII, and PIV was examined by real-time quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry in human synovial tissues, arthritic mouse joints, and human and murine FLS. CIA was induced in mice in which isoform PIV of Ciita was knocked out (PIV(-/-) ), in PIV(-/-) mice transgenic for CIITA in the thymus (K14 CIITA), and in their control littermates. RESULTS HLA-DRA, total CIITA, and CIITA PIII messenger RNA levels were significantly increased in synovial tissue samples from patients with rheumatoid arthritis compared with the levels in tissue from patients with osteoarthritis. Human FLS expressed surface class II MHC via CIITA PIII and PIV, while class II MHC expression in murine FLS was entirely mediated by PIV. Mice with a targeted deletion of CIITA PIV lack CD4+ T cells and were protected against CIA. The expression of CIITA was restored in the thymus of PIV(-/-) K14 CIITA-transgenic mice, which had a normal CD4+ T cell repertoire and normal surface levels of class II MHC on professional antigen-presenting cells, but did not induce class II MHC on FLS. Synovial inflammation and immune responses against type II collagen were similar in PIV(-/-) K14 CIITA-transgenic mice and control mice with CIA, but bone erosion was significantly reduced in the absence of PIV. CONCLUSION Overexpression of class II MHC is tightly correlated with CIITA expression in arthritic synovium and in FLS. Selective targeting of Ciita PIV in peripheral tissues abrogates class II MHC expression by murine FLS but does not protect against inflammation and autoimmune responses in CIA.
Collapse
|
41
|
Antigen recognition by autoreactive CD4⁺ thymocytes drives homeostasis of the thymic medulla. PLoS One 2012; 7:e52591. [PMID: 23300712 PMCID: PMC3531460 DOI: 10.1371/journal.pone.0052591] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/16/2012] [Indexed: 01/07/2023] Open
Abstract
The thymic medulla is dedicated for purging the T-cell receptor (TCR) repertoire of self-reactive specificities. Medullary thymic epithelial cells (mTECs) play a pivotal role in this process because they express numerous peripheral tissue-restricted self-antigens. Although it is well known that medulla formation depends on the development of single-positive (SP) thymocytes, the mechanisms underlying this requirement are incompletely understood. We demonstrate here that conventional SP CD4+ thymocytes bearing autoreactive TCRs drive a homeostatic process that fine-tunes medullary plasticity in adult mice by governing the expansion and patterning of the medulla. This process exhibits strict dependence on TCR-reactivity with self-antigens expressed by mTECs, as well as engagement of the CD28-CD80/CD86 costimulatory axis. These interactions induce the expression of lymphotoxin α in autoreactive CD4+ thymocytes and RANK in mTECs. Lymphotoxin in turn drives mTEC development in synergy with RANKL and CD40L. Our results show that Ag-dependent interactions between autoreactive CD4+ thymocytes and mTECs fine-tune homeostasis of the medulla by completing the signaling axes implicated in mTEC expansion and medullary organization.
Collapse
|
42
|
Pisapia L, Pozzo GD, Barba P, Citro A, Harris PE, Maffei A. Contrasting effects of IFNα on MHC class II expression in professional vs. nonprofessional APCs: Role of CIITA type IV promoter. RESULTS IN IMMUNOLOGY 2012; 2:174-83. [PMID: 24371581 DOI: 10.1016/j.rinim.2012.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/16/2012] [Accepted: 09/19/2012] [Indexed: 01/01/2023]
Abstract
We previously demonstrated that, in ex vivo cultures, IFNα downregulates the expression of MHC class II (MHCII) genes in human non-professional APCs associated with pancreatic islets. IFNα has an opposing effect on MHCII expression in professional APCs. In this study, we found that the mechanism responsible for the IFNα-mediated MHCII's downregulation in human MHCII-positive non-professional antigen presenting human non-hematopoietic cell lines is the result of the negative feedback system that regulates cytokine signal transduction, which eventually inhibits promoters III and IV of CIITA gene. Because the CIITA-PIV isoform is mostly responsible for the constitutive expression of MHCII genes in non-professional APCs, we pursued and achieved the specific knockdown of CIITA-PIV mRNA in our in vitro system, obtaining a partial silencing of MHCII molecules similar to that obtained by IFNα. We believe that our results offer a new understanding of the potential significance of CIITA-PIV as a therapeutic target for interventional strategies that can manage autoimmune disease and allograft rejection with little interference on the function of professional APCs of the immune system.
Collapse
Affiliation(s)
- Laura Pisapia
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Giovanna Del Pozzo
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Pasquale Barba
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Alessandra Citro
- Department of Medicine of Columbia University Medical Center, New York, NY, USA
| | - Paul E Harris
- Department of Medicine of Columbia University Medical Center, New York, NY, USA
| | - Antonella Maffei
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy ; Department of Medicine of Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
43
|
Yoon HS, Scharer CD, Majumder P, Davis CW, Butler R, Zinzow-Kramer W, Skountzou I, Koutsonanos DG, Ahmed R, Boss JM. ZBTB32 is an early repressor of the CIITA and MHC class II gene expression during B cell differentiation to plasma cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:2393-403. [PMID: 22851713 DOI: 10.4049/jimmunol.1103371] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CIITA and MHC class II expression is silenced during the differentiation of B cells to plasma cells. When B cell differentiation is carried out ex vivo, CIITA silencing occurs rapidly, but the factors contributing to this event are not known. ZBTB32, also known as repressor of GATA3, was identified as an early repressor of CIITA in an ex vivo plasma cell differentiation model. ZBTB32 activity occurred at a time when B lymphocyte-induced maturation protein-1 (Blimp-1), the regulator of plasma cell fate and suppressor of CIITA, was minimally induced. Ectopic expression of ZBTB32 suppressed CIITA and I-A gene expression in B cells. Short hairpin RNA depletion of ZBTB32 in a plasma cell line resulted in re-expression of CIITA and I-A. Compared with conditional Blimp-1 knockout and wild-type B cells, B cells from ZBTB32/ROG-knockout mice displayed delayed kinetics in silencing CIITA during ex vivo plasma cell differentiation. ZBTB32 was found to bind to the CIITA gene, suggesting that ZBTB32 directly regulates CIITA. Lastly, ZBTB32 and Blimp-1 coimmunoprecipitated, suggesting that the two repressors may ultimately function together to silence CIITA expression. These results introduce ZBTB32 as a novel regulator of MHC-II gene expression and a potential regulatory partner of Blimp-1 in repressing gene expression.
Collapse
Affiliation(s)
- Hye Suk Yoon
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
D'Agostino J, Ding X, Zhang P, Jia K, Fang C, Zhu Y, Spink DC, Zhang QY. Potential biological functions of cytochrome P450 reductase-dependent enzymes in small intestine: novel link to expression of major histocompatibility complex class II genes. J Biol Chem 2012; 287:17777-17788. [PMID: 22453923 DOI: 10.1074/jbc.m112.354274] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NADPH-cytochrome P450 reductase (POR) is essential for the functioning of microsomal cytochrome P450 (P450) monooxygenases and heme oxygenases. The biological roles of the POR-dependent enzymes in the intestine have not been defined, despite the wealth of knowledge on the biochemical properties of the various oxygenases. In this study, cDNA microarray analysis revealed significant changes in gene expression in enterocytes isolated from the small intestine of intestinal epithelium-specific Por knock-out (named IE-Cpr-null) mice compared with that observed in wild-type (WT) littermates. Gene ontology analyses revealed significant changes in terms related to P450s, transporters, cholesterol biosynthesis, and, unexpectedly, antigen presentation/processing. The genomic changes were confirmed at either mRNA or protein level for selected genes, including those of the major histocompatibility complex class II (MHC II). Cholesterol biosynthetic activity was greatly reduced in the enterocytes of the IE-Cpr-null mice, as evidenced by the accumulation of the lanosterol metabolite, 24-dihydrolanosterol. However, no differences in either circulating or enterocyte cholesterol levels were observed between IE-Cpr-null and WT mice. Interestingly, the levels of the cholesterol precursor farnesyl pyrophosphate and its derivative geranylgeranyl pyrophosphate were also increased in the enterocytes of the IE-Cpr-null mice. Furthermore, the expression of STAT1 (signal transducer and activator of transcription 1), a downstream target of geranylgeranyl pyrophosphate signaling, was enhanced. STAT1 is an activator of CIITA, the class II transactivator for MHC II expression; CIITA expression was concomitantly increased in IE-Cpr-null mice. Overall, these findings provide a novel and mechanistic link between POR-dependent enzymes and the expression of MHC II genes in the small intestine.
Collapse
Affiliation(s)
- Jaime D'Agostino
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York 12201-0509
| | - Xinxin Ding
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York 12201-0509
| | - Peng Zhang
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York 12201-0509
| | - Kunzhi Jia
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York 12201-0509
| | - Cheng Fang
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York 12201-0509
| | - Yi Zhu
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York 12201-0509
| | - David C Spink
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York 12201-0509
| | - Qing-Yu Zhang
- Wadsworth Center, New York State Department of Health, and School of Public Health, State University of New York, Albany, New York 12201-0509.
| |
Collapse
|
45
|
Dyggve H, Meri S, Spillmann T, Lohi H, Kennedy LJ, Speeti M. Evaluation of DLA promoters in Doberman hepatitis. ACTA ACUST UNITED AC 2012; 78:446-50. [PMID: 22077625 DOI: 10.1111/j.1399-0039.2011.01784.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Doberman hepatitis (DH) is associated with homozygous DLA-DRB1*00601/DQA1*00401/DQB1*01303 indicating a role for the immune system in the development of the disease. The dog leucocyte antigen (DLA) class II expression is controlled at the transcriptional level with proximal promoters. Differential expression of DLA class II molecules of antigen-presenting cells is reported to affect susceptibility to or protection from different immune-mediated diseases. The aim of this study was to evaluate, whether the variation in promoter areas of homozygous DLA-DRB1*00601/DQA1*00401/DQB1*01303 Dobermans could explain why some dogs become afflicted with DH and others do not. Our findings suggest that promoter variants are not associated as risk modifiers in homozygous DLA-DRB1*00601/DQA1*00401/DQB1*01303 Dobermans, but additional factors are needed. Nevertheless, our study indicates that the whole DLA block is associated to the disease.
Collapse
Affiliation(s)
- H Dyggve
- Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
46
|
CIITA promoter I CARD-deficient mice express functional MHC class II genes in myeloid and lymphoid compartments. Genes Immun 2012; 13:299-310. [PMID: 22218223 PMCID: PMC3366023 DOI: 10.1038/gene.2011.86] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Three distinct promoters control the master regulator of MHC class II expression, CIITA, in a cell type specific manner. Promoter I (pI) CIITA, expressed primarily by dendritic cells and macrophages, expresses a unique isoform that contains a caspase recruitment domain. The activity and function of this isoform is not understood but has been thought to enhance the function of CIITA in antigen presenting cells. To determine if isoform I of CIITA has specific functions, CIITA mutant mice were created in which isoform I was replaced with isoform III sequences. Mice in which pI and the CARD encoding exon were deleted were also created. No defect in the formation of CD4 T cells, the ability to respond to a model antigen, or bacterial or viral challenge was observed in mice lacking CIITA isoform I. Although CIITA and MHC-II expression was decreased in splenic DC, the pI knockout animals expressed CIITA from downstream promoters, suggesting that control of pI activity is mediated by unknown s II distal elements that could act at the pIII, the B cell promoter. Thus, no critical function is linked to the CARD domain of CIITA isoform I with respect to basic immune system development, function and challenge.
Collapse
|
47
|
Kumar P, Agarwal R, Siddiqui I, Vora H, Das G, Sharma P. ESAT6 differentially inhibits IFN‐γ‐inducible class II transactivator isoforms in both a TLR2‐dependent and ‐independent manner. Immunol Cell Biol 2011; 90:411-20. [DOI: 10.1038/icb.2011.54] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Pavanish Kumar
- Immunology Group, International Centre for Genetic Engineering and Biotechnology New Delhi India
| | - Richa Agarwal
- Immunology Group, International Centre for Genetic Engineering and Biotechnology New Delhi India
| | - Imran Siddiqui
- Immunology Group, International Centre for Genetic Engineering and Biotechnology New Delhi India
| | - Hardeep Vora
- Immunology Group, International Centre for Genetic Engineering and Biotechnology New Delhi India
| | - Gobardhan Das
- Immunology Group, International Centre for Genetic Engineering and Biotechnology New Delhi India
| | - Pawan Sharma
- Immunology Group, International Centre for Genetic Engineering and Biotechnology New Delhi India
| |
Collapse
|
48
|
Lee AW, Wang N, Hornell TMC, Harding JJ, Deshpande C, Hertel L, Lacaille V, Pashine A, Macaubas C, Mocarski ES, Mellins ED. Human cytomegalovirus decreases constitutive transcription of MHC class II genes in mature Langerhans cells by reducing CIITA transcript levels. Mol Immunol 2011; 48:1160-7. [PMID: 21458073 DOI: 10.1016/j.molimm.2011.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 02/20/2011] [Accepted: 02/21/2011] [Indexed: 11/29/2022]
Abstract
Human cytomegalovirus (HCMV) productively infects CD34(+) progenitor-derived, mature Langerhans-type dendritic cells (matLC) and reduces surface expression of MHC class II complexes (MHC II) by increasing intracellular retention of these molecules. To determine whether HCMV also inhibits MHC II expression by other mechanisms, we assessed mRNA levels of the class II transcriptional regulator, CIITA, and several of its target genes in infected matLC. Levels of CIITA, HLA-DRA (DRA) and DRB transcripts, and new DR protein synthesis were compared in mock-infected and HCMV-infected cells by quantitative PCR and pulse-chase immunoprecipitation analyses, respectively. CIITA mRNA levels were significantly lower in HCMV-infected matLC as compared to mock-infected cells. When assessed in the presence of Actinomycin D, the stability of CIITA transcripts was not diminished by HCMV. Analysis of promoter-specific CIITA isoforms revealed that types I, III and IV all were decreased by HCMV, a result that differs from changes after incubation of these cells with lipopolysaccharide (LPS). Exposure to UV-inactivated virus failed to reduce CIITA mRNA levels, implicating de novo viral gene expression in this effect. HCMV-infected matLC also expressed lower levels of DR transcripts and reduced DR protein synthesis rates compared to mock-infected matLC. In summary, we demonstrate that HCMV infection of a human dendritic cell subset inhibits constitutive CIITA expression, most likely at the transcriptional level, resulting in reduced MHC II biosynthesis. We suggest this represents a new mechanism of modulation of mature LC by HCMV.
Collapse
Affiliation(s)
- Andrew W Lee
- Department of Pediatrics, Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kim EJ, Choi B, Moon H, Lee YJ, Jeon YK, Park SH, Kim TJ, Jung KC. CD4(+) T cells from MHC II-dependent thymocyte-thymocyte interaction provide efficient help for B cells. Immunol Cell Biol 2011; 89:897-903. [PMID: 21358747 PMCID: PMC3257047 DOI: 10.1038/icb.2011.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recently, a novel CD4+ T-cell developmental pathway was reported that generates thymocyte–thymocyte (T–T) CD4+ T cells. We established a mouse system (CIITAtgCIITApIV−/−) where thymic positive selection occurred only by major histocompatibility complex (MHC) class II+ thymocytes. T–T CD4+ T cells selected via MHC class II-dependent T–T interaction are comprised of PLZF-negative and innate PLZF-positive populations. Until recently, the functional role of the PLZF-negative population was unclear. In this study, we demonstrate that naïve T–T CD4+ T cells provide B-cell help to a level comparable with that of naïve conventional CD4+ T cells. Considering the absence of PLZF expression in naïve T–T CD4+ T cells, these results suggest that PLZF-negative naïve T–T CD4+ T cells are functionally equivalent to conventional naïve CD4+ T cells in terms of B-cell help.
Collapse
Affiliation(s)
- Eun Ji Kim
- Graduate School of Immunology, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Smith MA, Wright G, Wu J, Tailor P, Ozato K, Chen X, Wei S, Piskurich JF, Ting JPY, Wright KL. Positive regulatory domain I (PRDM1) and IRF8/PU.1 counter-regulate MHC class II transactivator (CIITA) expression during dendritic cell maturation. J Biol Chem 2011; 286:7893-7904. [PMID: 21216962 DOI: 10.1074/jbc.m110.165431] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) are key mediators of immune function through robust and tightly regulated presentation of antigen in the context of the MHC Class II. MHC Class II expression is controlled by the transactivator CIITA. CIITA expression in conventional DCs is uniquely dependent on an uncharacterized myeloid cell-specific promoter, CIITApI. We now identify in vivo the promoter structure and factors regulating CIITApI. In immature DCs transcription requires binding of PU.1, IRF8, NFκB, and Sp1 to the promoter. PU.1 binds independently at one site and in a required heterodimer with IRF8 at a composite element. DCs from IRF8-null mice have an unoccupied CIITApI promoter that can be rescued by reconstitution with IRF8 in vitro. Furthermore, mutation of either PU.1 site or the IFR8 site inhibits transcriptional activation. In vivo footprinting and chromatin immunoprecipitation reveals that DC maturation induces complete disassociation of the bound activators paralleled by recruitment of PRDM1/Blimp-1 to the promoter. PRDM1 is a transcriptional repressor with essential roles in B cells, T cells, NK cells, and DCs. We show that PRDM1 co-repressors, G9a and HDAC2, are recruited to CIITApI, leading to a loss of histone acetylation and acquisition of histone H3K9 dimethylation and heterochromatin protein 1γ (HP1γ). PRDM1 binding also blocks IRF8-mediated activation dependent on the PU.1/IRF composite element. Together these findings reveal the mechanisms regulating CIITA and, thus, antigen presentation in DCs, demonstrating that PRDM1 and IRF8/PU.1 counter-regulate expression. The activity of PRDM1 in silencing all three cell type-specific CIITA promoters places it as a central regulator of antigen presentation.
Collapse
Affiliation(s)
- Matthew A Smith
- From the H. Lee Moffitt Cancer Center and Research Institute, Department of Molecular Medicine and Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33612
| | - Gabriela Wright
- From the H. Lee Moffitt Cancer Center and Research Institute, Department of Molecular Medicine and Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33612
| | - Jian Wu
- From the H. Lee Moffitt Cancer Center and Research Institute, Department of Molecular Medicine and Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33612
| | - Prafullakumar Tailor
- the Laboratory of Molecular Growth Regulation, NICHD, National Institutes of Health, Bethesda, Maryland 20892
| | - Keiko Ozato
- the Laboratory of Molecular Growth Regulation, NICHD, National Institutes of Health, Bethesda, Maryland 20892
| | - Xianghong Chen
- From the H. Lee Moffitt Cancer Center and Research Institute, Department of Molecular Medicine and Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33612
| | - Sheng Wei
- From the H. Lee Moffitt Cancer Center and Research Institute, Department of Molecular Medicine and Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33612
| | - Janet F Piskurich
- the Department of Medical Education, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas 79905, and
| | - Jenny P-Y Ting
- the Department of Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Kenneth L Wright
- From the H. Lee Moffitt Cancer Center and Research Institute, Department of Molecular Medicine and Department of Oncologic Sciences, University of South Florida, Tampa, Florida 33612,.
| |
Collapse
|