1
|
Chandwaskar R, Dalal R, Gupta S, Sharma A, Parashar D, Kashyap VK, Sohal JS, Tripathi SK. Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease. Scand J Immunol 2024; 100:e13412. [PMID: 39394898 DOI: 10.1111/sji.13412] [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: 11/20/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Inflammatory bowel disease (IBD), comprised of Crohn's disease (CD) and ulcerative colitis (UC), are gut inflammatory diseases that were earlier prevalent in the Western Hemisphere but now are on the rise in the East, with India standing second highest in the incidence rate in the world. Inflammation in IBD is a cause of dysregulated immune response, wherein helper T (Th) cell subsets and their cytokines play a major role in the pathogenesis of IBD. In addition, gut microbiota, environmental factors such as dietary factors and host genetics influence the outcome and severity of IBD. Dysregulation between effector and regulatory T cells drives gut inflammation, as effector T cells like Th1, Th17 and Th9 subsets Th cell lineages were found to be increased in IBD patients. In this review, we attempted to discuss the role of different Th cell subsets together with other T cells like CD8+ T cells, NKT and γδT cells in the outcome of gut inflammation in IBD. We also highlighted the potential therapeutic candidates for IBD.
Collapse
Affiliation(s)
- Rucha Chandwaskar
- Amity Institute of Microbial Technology (AIMT), Amity University Jaipur, Rajasthan, India
| | - Rajdeep Dalal
- Infection and Immunology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, India
| | - Saurabh Gupta
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur, Karnataka, India
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Jagdip Singh Sohal
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Subhash K Tripathi
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
2
|
Vaillant L, Akhter W, Nakhle J, Simon M, Villalba M, Jorgensen C, Vignais ML, Hernandez J. The role of mitochondrial transfer in the suppression of CD8 + T cell responses by Mesenchymal stem cells. Stem Cell Res Ther 2024; 15:394. [PMID: 39497203 PMCID: PMC11536934 DOI: 10.1186/s13287-024-03980-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/04/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND . CD8+ Cytotoxic T lymphocytes play a key role in the pathogenesis of autoimmune diseases and clinical conditions such as graft versus host disease and graft rejection. Mesenchymal Stromal Cells (MSCs) are multipotent cells with tissue repair and immunomodulatory capabilities. Since they are able to suppress multiple pathogenic immune responses, MSCs have been proposed as a cellular therapy for the treatment of immune-mediated diseases. However, the mechanisms underlying their immunosuppressive properties are not yet fully understood. MSCs have the remarkable ability to sense tissue injury and inflammation and respond by donating their own mitochondria to neighboring cells. Whether mitochondrial transfer has any role in the repression of CD8+ responses is unknown. METHODS AND RESULTS . We have utilized CD8+ T cells from Clone 4 TCR transgenic mice that differentiate into effector cells upon activation in vitro and in vivo to address this question. Allogeneic bone marrow derived MSCs, co-cultured with activated Clone 4 CD8+ T cells, decreased their expansion, the production of the effector cytokine IFNγ and their diabetogenic potential in vivo. Notably, we found that during this interaction leading to suppression, MSCs transferred mitochondria to CD8+ T cells as evidenced by FACS and confocal microscopy. Transfer of MSC mitochondria to Clone 4 CD8+ T cells also resulted in decreased expansion and production of IFNγ upon activation. These effects overlapped and were additive with those of prostaglandin E2 secreted by MSCs. Furthermore, preventing mitochondrial transfer in co-cultures diminished the ability of MSCs to inhibit IFNγ production. Finally, we demonstrated that both MSCs and MSC mitochondria downregulated T-bet and Eomes expression, key transcription factors for CTL differentiation, on activated CD8+ T cells. CONCLUSION . In this report we showed that MSCs are able to interact with CD8+ T cells and transfer them their mitochondria. Mitochondrial transfer contributed to the global suppressive effect of MSCs on CD8+ T cell activation by downregulating T-bet and Eomes expression resulting in impaired IFNγ production of activated CD8+ T cells.
Collapse
Affiliation(s)
- Loic Vaillant
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Waseem Akhter
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Jean Nakhle
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
- IGMM, Université de Montpellier, CNRS, Montpellier, France
| | - Matthieu Simon
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Martin Villalba
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Christian Jorgensen
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- CHU Montpellier, Montpellier, France
| | - Marie-Luce Vignais
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Javier Hernandez
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France.
| |
Collapse
|
3
|
Houen G. Auto-immuno-deficiency syndromes. Autoimmun Rev 2024; 23:103610. [PMID: 39209011 DOI: 10.1016/j.autrev.2024.103610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Autoimmune diseases constitute a broad, heterogenous group with many diverse and often overlapping symptoms. Even so, they are traditionally classified as either systemic, rheumatic diseases or organ-directed diseases. Several theories exist about autoimmune diseases, including defective self-recognition, altered self, molecular mimicry, bystander activation and epitope spreading. While there is no consensus about these theories, it is generally accepted that genetic, pre-disposing factors in combination with environmental factors can result in autoimmune disease. The relative contribution of genetic and environmental factors varies between diseases, as does the significance of individual contributing factors within related diseases. Among the genetic factors, molecules involved in antigen (Ag) recognition, processing, and presentation stand out (e.g., MHC I and II) together with molecules involved in immune signaling and regulation of cellular interactions (i.e., immuno-phenotypes). Also, various immuno-deficiencies have been linked to development of autoimmune diseases. Among the environmental factors, infections (e.g., viruses) have attracted most attention, but factors modulating the immune system have also been the subject of much research (e.g., sunlight and vitamin D). Multiple sclerosis currently stands out due to a very strong and proven association with Epstein-Barr virus infection, notably in cases of late infection and in cases of EBV-associated mononucleosis. Thus, a common picture is emerging that both systemic and organ-directed autoimmune diseases may appropriately be described as auto-immuno-deficiency syndromes (AIdeSs), a concept that emphasizes and integrates existing knowledge on the role of immuno-deficiencies and chronic infections with development of overlapping disease syndromes with variable frequencies of autoantibodies and/or autoreactive T cells. This review integrates and exemplifies current knowledge on the interplay of genetically determined immuno-phenotypes and chronic infections in the development of AIdeSs.
Collapse
Affiliation(s)
- Gunnar Houen
- Department of Neurology and Translational Research Center (TRACE), Rigshospitalet, Nordstjernevej 42, DK-2600 Glostrup, Denmark; Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark.
| |
Collapse
|
4
|
Padoan B, Casar C, Krause J, Schultheiss C, Baumdick ME, Niehrs A, Zecher BF, Pujantell M, Yuki Y, Martin M, Remmerswaal EBM, Dekker T, van der Bom-Baylon ND, Noble JA, Carrington M, Bemelman FJ, van Lier RAW, Binder M, Gagliani N, Bunders MJ, Altfeld M. NKp44/HLA-DP-dependent regulation of CD8 effector T cells by NK cells. Cell Rep 2024; 43:114089. [PMID: 38615318 PMCID: PMC11416720 DOI: 10.1016/j.celrep.2024.114089] [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: 10/30/2023] [Revised: 03/03/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Although natural killer (NK) cells are recognized for their modulation of immune responses, the mechanisms by which human NK cells mediate immune regulation are unclear. Here, we report that expression of human leukocyte antigen (HLA)-DP, a ligand for the activating NK cell receptor NKp44, is significantly upregulated on CD8+ effector T cells, in particular in human cytomegalovirus (HCMV)+ individuals. HLA-DP+ CD8+ T cells expressing NKp44-binding HLA-DP antigens activate NKp44+ NK cells, while HLA-DP+ CD8+ T cells not expressing NKp44-binding HLA-DP antigens do not. In line with this, frequencies of HLA-DP+ CD8+ T cells are increased in individuals not encoding for NKp44-binding HLA-DP haplotypes, and contain hyper-expanded CD8+ T cell clones, compared to individuals expressing NKp44-binding HLA-DP molecules. These findings identify a molecular interaction facilitating the HLA-DP haplotype-specific editing of HLA-DP+ CD8+ T cell effector populations by NKp44+ NK cells and preventing the generation of hyper-expanded T cell clones, which have been suggested to have increased potential for autoimmunity.
Collapse
Affiliation(s)
- Benedetta Padoan
- Research Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Christian Casar
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jenny Krause
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Christoph Schultheiss
- Division of Medical Oncology, University Hospital Basel, 4031 Basel, Switzerland; Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, 4031 Basel, Switzerland
| | - Martin E Baumdick
- Research Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Annika Niehrs
- Research Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Britta F Zecher
- Research Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany; I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Pujantell
- Research Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Yuko Yuki
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Maureen Martin
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Tamara Dekker
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Nelly D van der Bom-Baylon
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Janelle A Noble
- Department of Pediatrics UCSF, Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Mary Carrington
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Frederike J Bemelman
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, 4031 Basel, Switzerland; Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, 4031 Basel, Switzerland
| | - Nicola Gagliani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
| | - Madeleine J Bunders
- Research Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
| | - Marcus Altfeld
- Research Department Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany; Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany.
| |
Collapse
|
5
|
Aggio V, Fabbella L, Poletti S, Lorenzi C, Finardi A, Colombo C, Zanardi R, Furlan R, Benedetti F. Circulating cytotoxic immune cell composition, activation status and toxins expression associate with white matter microstructure in bipolar disorder. Sci Rep 2023; 13:22209. [PMID: 38097657 PMCID: PMC10721611 DOI: 10.1038/s41598-023-49146-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023] Open
Abstract
Patients with bipolar disorder (BD) show higher immuno-inflammatory setpoints, with in vivo alterations in white matter (WM) microstructure and post-mortem infiltration of T cells in the brain. Cytotoxic CD8+ T cells can enter and damage the brain in inflammatory disorders, but little is known in BD. Our study aimed to investigate the relationship between cytotoxic T cells and WM alterations in BD. In a sample of 83 inpatients with BD in an active phase of illness (68 depressive, 15 manic), we performed flow cytometry immunophenotyping to investigate frequencies, activation status, and expression of cytotoxic markers in CD8+ and tested for their association with diffusion tensor imaging (DTI) measures of WM microstructure. Frequencies of naïve and activated CD8+ cell populations expressing Perforin, or both Perforin and Granzyme, negatively associated with WM microstructure. CD8+ Naïve cells negative for Granzyme and Perforin positively associates with indexes of WM integrity, while the frequency of CD8+ memory cells negatively associates with index of WM microstructure, irrespective of toxins expression. The resulting associations involve measures representative of orientational coherence and myelination of the fibers (FA and RD), suggesting disrupted oligodendrocyte-mediated myelination. These findings seems to support the hypothesis that immunosenescence (less naïve, more memory T cells) can detrimentally influence WM microstructure in BD and that peripheral CD8+ T cells may participate in inducing an immune-related WM damage in BD mediated by killer proteins.
Collapse
Affiliation(s)
- Veronica Aggio
- Psychiatry and Clinical Psychobiology Unit, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, San Raffaele Turro, Via Stamira d'Ancona 20, 20127, Milano, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Lorena Fabbella
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Poletti
- Psychiatry and Clinical Psychobiology Unit, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, San Raffaele Turro, Via Stamira d'Ancona 20, 20127, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology Unit, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, San Raffaele Turro, Via Stamira d'Ancona 20, 20127, Milano, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Colombo
- Vita-Salute San Raffaele University, Milan, Italy
- Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Raffaella Zanardi
- Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Roberto Furlan
- Vita-Salute San Raffaele University, Milan, Italy
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Benedetti
- Psychiatry and Clinical Psychobiology Unit, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, San Raffaele Turro, Via Stamira d'Ancona 20, 20127, Milano, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
6
|
Lee JS, Karthikeyan D, Fini M, Vincent BG, Rubinsteyn A. ACE configurator for ELISpot: optimizing combinatorial design of pooled ELISpot assays with an epitope similarity model. Brief Bioinform 2023; 25:bbad495. [PMID: 38180831 PMCID: PMC10768796 DOI: 10.1093/bib/bbad495] [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: 08/22/2023] [Revised: 11/16/2023] [Accepted: 12/01/2023] [Indexed: 01/07/2024] Open
Abstract
The enzyme-linked immunosorbent spot (ELISpot) assay is a powerful in vitro immunoassay that enables cost-effective quantification of antigen-specific T-cell reactivity. It is used widely in the context of cancer and infectious diseases to validate the immunogenicity of predicted epitopes. While technological advances have kept pace with the demand for increased throughput, efforts to increase scale are bottlenecked by current assay design and deconvolution methods, which have remained largely unchanged. Current methods for designing pooled ELISpot experiments offer limited flexibility of assay parameters, lack support for high-throughput scenarios and do not consider peptide identity during pool assignment. We introduce the ACE Configurator for ELISpot (ACE) to address these gaps. ACE generates optimized peptide-pool assignments from highly customizable user inputs and handles the deconvolution of positive peptides using assay readouts. In this study, we present a novel sequence-aware pooling strategy, powered by a fine-tuned ESM-2 model that groups immunologically similar peptides, reducing the number of false positives and subsequent confirmatory assays compared to existing combinatorial approaches. To validate ACE's performance on real-world datasets, we conducted a comprehensive benchmark study, contextualizing design choices with their impact on prediction quality. Our results demonstrate ACE's capacity to further increase precision of identified immunogenic peptides, directly optimizing experimental efficiency. ACE is freely available as an executable with a graphical user interface and command-line interfaces at https://github.com/pirl-unc/ace.
Collapse
Affiliation(s)
- Jin Seok Lee
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Dhuvarakesh Karthikeyan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Misha Fini
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Alex Rubinsteyn
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| |
Collapse
|
7
|
Dai YW, Wang WM, Zhou X. Development of a CD8 + T cell-based molecular classification for predicting prognosis and heterogeneity in triple-negative breast cancer by integrated analysis of single-cell and bulk RNA-sequencing. Heliyon 2023; 9:e19798. [PMID: 37810147 PMCID: PMC10559128 DOI: 10.1016/j.heliyon.2023.e19798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC), although the most intractable subtype, is characterized by abundant immunogenicity, which enhances responsiveness to immunotherapeutic measures. Methods First, we identified CD8+ T cell core genes (TRCG) based on single-cell sequence and traditional transcriptome sequencing and then used this data to develop a first-of-its-kind classification system based on CD8+ T cells in patients with TNBC. Next, TRCG-related patterns were systematically analyzed, and their correlation with genomic features, immune activity (microenvironment associated with immune infiltration), and clinicopathological characteristics were assessed in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC), the Cancer Genome Atlas (TCGA), GSE103091, GSE96058 databases. Additionally, a CD8+ T cell-related prognostic signature (TRPS) was developed to quantify a patient-specific TRCG pattern. What's more, the genes-related TRPS was validated by polymerase chain reaction (PCR) experiment. Results This study, for the first time, distinguished two subsets in patients with TNBC based on the TRCG. The immune microenvironment and prognostic stratification between these have distinct heterogeneity. Furthermore, this study constructed a novel scoring system named TRPS, which we show to be a robust prognostic marker for TNBC that is related to the intensity of immune infiltration and immunotherapy. Moreover, the levels of genes related the TRPS were validated by quantitative Real-Time PCR. Conclusions Consequently, this study unraveled an association between the TRCG and the tumor microenvironment in TNBC. TRPS model represents an effective tool for survival prediction and treatment guidance in TNBC that can also help identify individual variations in TME and stratify patients who are sensitive to anticancer immunotherapy.
Collapse
Affiliation(s)
- Yin-wei Dai
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Wei-ming Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiang Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, China
| |
Collapse
|
8
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 266] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
9
|
Moreno-Gonzalez MA, Ortega-Rivera OA, Steinmetz NF. Two decades of vaccine development against atherosclerosis. NANO TODAY 2023; 50:101822. [PMID: 37860053 PMCID: PMC10586238 DOI: 10.1016/j.nantod.2023.101822] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Atherosclerosis is an immune-mediated chronic inflammatory disease that leads to the development of fatty plaques in the arterial walls, ultimately increasing the risk of thrombosis, stroke, and myocardial infarction. The immune response in this complex disease is both atheroprotective and pro-atherogenic, involving both innate and adaptive immunity. Current treatments include the adjustment of lifestyle factors, cholesterol-lowering drugs such as statins, and immunotherapy, whereas vaccine development has received comparatively little attention. In this review, we discuss the potential of antigen-specific vaccination as a preventative approach based on more than 20 years of research and innovation. Vaccination targets include proteins that are more abundant in atherosclerotic patients, such as oxidized low-density lipoprotein (LDL), apolipoprotein B-100, proprotein convertase subtilisin/kexin type-9 serine protease (PCSK9), cholesteryl ester transfer protein (CETP), and heat shock proteins HSP60 and HSP65. Immunization with such proteins or their peptide epitopes has been shown to induce T-cell activation, produce antigen-specific antibodies, reduce the size of atherosclerotic lesions, and/or reduce serum cholesterol levels. Vaccination against atherosclerosis therefore offers a new strategy to address the burden on healthcare systems caused by cardiovascular disease, the leading cause of death worldwide.
Collapse
Affiliation(s)
- Miguel A. Moreno-Gonzalez
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
| | - Oscar A. Ortega-Rivera
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Institute for Materials Discovery and Design, University of California-San Diego, La Jolla, CA 92039, USA
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92039, USA
- Department of Radiology, University of California-San Diego, La Jolla, CA 92039, USA
- Moores Cancer Center, University of California-San Diego, La Jolla, CA 92039, USA
| |
Collapse
|
10
|
Shan J, Li L, Du L, Yang P. Association of TBX21 gene polymorphisms and acute anterior uveitis risk in a Chinese population: A case-control study. Exp Eye Res 2023; 229:109417. [PMID: 36796535 DOI: 10.1016/j.exer.2023.109417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/27/2022] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Copy number variations (CNVs) in TBX21 gene have been reported to be significantly and positively correlated with acute anterior uveitis (AAU). Our study was performed to further determine whether single nucleotide polymorphisms (SNPs) in TBX21 gene confer susceptibility to AAU in a Chinese population. In our case-control study, 420 AAU patients and 918 healthy controls were included. SNP genotyping was conducted via the MassARRAY™ iPLEX Gold platform. Association and haplotype analyses were performed via SPSS 23.0 and SHEsis software. No significant association was observed between two candidate SNPs of TBX21 gene (rs4794067, rs11657479) and susceptibility to AAU (Pc > 0.05). In stratification analysis, the result also showed no significant difference between the HLA-B27 positive AAU patients and non-typed healthy controls. Additionally, no association was detected between TBX21 haplotypes and AAU risk. In conclusion, the polymorphisms rs4794067 and rs11657479 in TBX21 gene did not confer disease susceptibility to AAU in a Chinese population.
Collapse
Affiliation(s)
- Jiankang Shan
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, People's Republic of China; Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, People's Republic of China
| | - Lin Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, People's Republic of China
| | - Liping Du
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, People's Republic of China.
| | - Peizeng Yang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, People's Republic of China; The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing Branch (Municipal Division) of National Clinical Research Center for Ocular Diseases, Chongqing, People's Republic of China.
| |
Collapse
|
11
|
Chromogranin A: An Endocrine Factor of Pregnancy. Int J Mol Sci 2023; 24:ijms24054986. [PMID: 36902417 PMCID: PMC10002927 DOI: 10.3390/ijms24054986] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/26/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Pregnancy is a state of physiological and hormonal changes. One of the endocrine factors involved in these processes is chromogranin A, an acidic protein produced, among others, by the placenta. Although it has been previously linked to pregnancy, no existing articles have ever managed to clarify the role of this protein regarding this subject. Therefore, the aim of the present study is to gather knowledge of chromogranin A's function with reference to gestation and parturition, clarify elusive information, and, most importantly, to formulate hypotheses for the future studies to verify.
Collapse
|
12
|
Li C, Guan N, Liu F. T7 peptide-decorated exosome-based nanocarrier system for delivery of Galectin-9 siRNA to stimulate macrophage repolarization in glioblastoma. J Neurooncol 2023; 162:93-108. [PMID: 36854924 DOI: 10.1007/s11060-023-04257-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Exosomes are nano-vesicular carriers capable of delivering cargoes for intercellular communication, which holds potential as biocompatible and high efficiency systems for drug delivery. In this study, we evaluated the potential effect of T7 peptide-decorated exosome-loaded Galectin-9 siRNA (T7-Exo/siGalectin-9) in the M1 polarization of macrophages and immunosuppression of glioblastoma (GBM). METHODS Differentially expressed genes in GBM were in silico predicted and then experimentally verified. Galectin-9 was knocked down by siRNA to assess its role in tumor-bearing mice. T7 peptide-decorated exosomes (derived from human embryonic kidney [HEK]-293T cells) targeting GBM were prepared, and loaded with Galectin-9 siRNA by electroporation to prepare nanoformulations (T7-Exo/siGalectin-9). The role of T7-Exo/siGalectin-9 in CD8+ T cell cytotoxicity to target GBM cells and polarization of macrophages was evaluated after artificial modulation of Galectin-9 expression. Anti-tumor effects of T7-Exo/siGalectin-9 were elucidated in vitro and in vivo. RESULTS Galectin-9 was highly expressed in GBM tissues and cell lines. The siRNA-mediated knockdown of Galectin-9 repressed the growth of xenografts of GBM cells in C57BL/6 mice and activated immune response in the tumor microenvironment. T7-Exo/siGalectin-9 effectively delivered siGalectin-9 to GBM cells. T7-Exo/siGalectin-9 contributed to activation of the TLR7-IRF5 pathway, which polarized macrophages to M1 phenotype. By this mechanism, phagocytosis of GBM cells by macrophages was increased, the anti-tumor effect of CD8+ T cells was enhanced and the inflammatory responses were suppressed. CONCLUSION Overall, T7-Exo/siGalectin-9 promotes macrophage repolarization and restricts the immunosuppression of GBM, thus providing novel insights into and drug delivery system of immunotherapy for GBM.
Collapse
Affiliation(s)
- Chenguang Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Ning Guan
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121000, Liaoning, People's Republic of China.
| |
Collapse
|
13
|
Li Y, Wang Q, Li J, Li A, Wang Q, Zhang Q, Chen Y. Therapeutic modulation of V Set and Ig domain-containing 4 (VSIG4) signaling in immune and inflammatory diseases. Cytotherapy 2023; 25:561-572. [PMID: 36642683 DOI: 10.1016/j.jcyt.2022.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023]
Abstract
Inflammation is the result of acute and chronic stresses, caused by emotional or physical trauma, or nutritional or environmental pollutants, and brings serious harm to human life and health. As an important cellular component of the innate immune barrier, the macrophage plays a key role in maintaining tissue homeostasis and promoting tissue repair by controlling infection and resolving inflammation. Several studies suggest that V Set and Ig domain-containing 4 is specifically expressed in tissue macrophages and is associated with a variety of inflammatory diseases. In this paper, we mainly summarize the recent research on V Set and Ig domain-containing 4 structures, functions, function and roles in acute and chronic inflammatory diseases, and provide a novel therapeutic avenue for the treatment of inflammatory diseases, including nervous system, urinary, respiratory and metabolic diseases.
Collapse
Affiliation(s)
- You Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Qi Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Jiaxin Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Aohan Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Qianqian Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China.
| | - Yingqing Chen
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China; Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, Liaoning, China.
| |
Collapse
|
14
|
Harding D, Chong MHA, Lahoti N, Bigogno CM, Prema R, Mohiddin SA, Marelli-Berg F. Dilated cardiomyopathy and chronic cardiac inflammation: Pathogenesis, diagnosis and therapy. J Intern Med 2023; 293:23-47. [PMID: 36030368 DOI: 10.1111/joim.13556] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dilated cardiomyopathy (DCM) is typically defined by left ventricular dilation and systolic dysfunction in the absence of a clear precipitant. Idiopathic disease is common; up to 50% of patients with DCM have no cause found despite imaging, genetic and biopsy assessments. Treatment remains focused on managing symptoms, reducing the risk of sudden cardiac death and ameliorating the structural and electrical complications of disease progression. In the absence of aetiology-specific treatments, the condition remains associated with a poor prognosis; mortality is approximately 40% at 10 years. The role of immune-mediated inflammatory injury in the development and progression of DCM was first proposed over 30 years ago. Despite the subsequent failures of three large clinical trials of immunosuppressive treatment (ATTACH, RENEWAL and the Myocarditis Treatment Trial), evidence for an abnormal adaptive immune response in DCM remains significant. In this review, we summarise and discuss available evidence supporting immune dysfunction in DCM, with a specific focus on cellular immunity. We also highlight current clinical and experimental treatments. We propose that the success of future immunosuppressive treatment trials in DCM will be dependent on the deep immunophenotyping of patients, to identify those with active inflammation and/or an abnormal immune response who are most likely to respond to therapy.
Collapse
Affiliation(s)
- Daniel Harding
- Centre for Biochemical Pharmacology, William Harvey Research Institute, London, UK
| | - Ming H A Chong
- Barts and The London School of Medicine and Dentistry, London, UK
| | - Nishant Lahoti
- Conquest Hospital, East Sussex Healthcare NHS Trust, St Leonards-on-Sea, UK
| | - Carola M Bigogno
- Barts and The London School of Medicine and Dentistry, London, UK
| | - Roshni Prema
- University Hospital, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | | | | |
Collapse
|
15
|
Radziszewska A, Moulder Z, Jury EC, Ciurtin C. CD8 + T Cell Phenotype and Function in Childhood and Adult-Onset Connective Tissue Disease. Int J Mol Sci 2022; 23:11431. [PMID: 36232733 PMCID: PMC9569696 DOI: 10.3390/ijms231911431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
CD8+ T cells are cytotoxic lymphocytes that destroy pathogen infected and malignant cells through release of cytolytic molecules and proinflammatory cytokines. Although the role of CD8+ T cells in connective tissue diseases (CTDs) has not been explored as thoroughly as that of other immune cells, research focusing on this key component of the immune system has recently gained momentum. Aberrations in cytotoxic cell function may have implications in triggering autoimmunity and may promote tissue damage leading to exacerbation of disease. In this comprehensive review of current literature, we examine the role of CD8+ T cells in systemic lupus erythematosus, Sjögren's syndrome, systemic sclerosis, polymyositis, and dermatomyositis with specific focus on comparing what is known about CD8+ T cell peripheral blood phenotypes, CD8+ T cell function, and CD8+ T cell organ-specific profiles in adult and juvenile forms of these disorders. Although, the precise role of CD8+ T cells in the initiation of autoimmunity and disease progression remains to be elucidated, increasing evidence indicates that CD8+ T cells are emerging as an attractive target for therapy in CTDs.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Zachary Moulder
- University College London Medical School, University College London, London WC1E 6DE, UK
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| |
Collapse
|
16
|
Pani F, Caria P, Yasuda Y, Makoto M, Mariotti S, Leenhardt L, Roshanmehr S, Caturegli P, Buffet C. The Immune Landscape of Papillary Thyroid Cancer in the Context of Autoimmune Thyroiditis. Cancers (Basel) 2022; 14:cancers14174287. [PMID: 36077831 PMCID: PMC9454449 DOI: 10.3390/cancers14174287] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The association between papillary thyroid cancer and Hashimoto’s thyroiditis went through a long-standing human debate recently elucidated by the establishment of a novel mouse model. Papillary thyroid carcinoma is an excellent model for studying the tumor immune microenvironment because it is naturally accompanied by immune cells, making it a good candidate for the treatment with immune checkpoint inhibitors. Abstract Papillary thyroid cancer (PTC) often co-occurs with Hashimoto’s thyroiditis, an association that has long been reported in clinical studies, remaining controversial. Experimental evidence has recently shown that pre-existing thyroiditis has a beneficial effect on PTC growth and progression by a distinctive expansion of effector memory CD8 T cells. Although the link between inflammation and PTC might involve different components of the immune system, a deep characterization of them which includes T cells, B cells and tertiary lymphoid structures, Mye-loid cells, Neutrophils, NK cells and dendritic cells will be desirable. The present review article considers the role of the adaptive and innate immune response surrounding PTC in the context of Hashimoto’s thyroiditis. This review will focus on the current knowledge by in vivo and in vitro studies specifically performed on animals’ models; thyroid cancer cells and human samples including (i) the dual role of tumor-infiltrating lymphocytes; (ii) the emerging role of B cells and tertiary lymphoid structures; (iii) the role of myeloid cells, dendritic cells, and natural killer cells; (iv) the current knowledge of the molecular biomarkers implicated in the complex link between thyroiditis and PTC and the potential implication of cancer immunotherapy in PTC patients in the context of thyroiditis.
Collapse
Affiliation(s)
- Fabiana Pani
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
- Correspondence: or
| | - Paola Caria
- Department of Biomedical Sciences, Biochemistry, Biology and Genetics Unit, University of Cagliari, Cittadella Universitaria di Monserrato, SP 8, Km 0.700, Monserrato, 09042 Cagliari, Italy
| | - Yoshinori Yasuda
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Miyara Makoto
- Inserm, Centre d’Immunologie et des Maladies Infectieuses-Paris (CIMI-PARIS), AP-HP Hôpital Pitié-Salpêtrière, Sorbonne Université, 75013 Paris, France
| | - Stefano Mariotti
- Department of Medical Sciences and Public Health, Endocrinology Unit, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | - Laurence Leenhardt
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
| | - Solmaz Roshanmehr
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Patrizio Caturegli
- Division of Immunology, Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Camille Buffet
- Service des Pathologies Thyroïdiennes et Tumeurs Endocrines, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, GRC n°16, GRC Tumeurs Thyroïdiennes, 75013 Paris, France
| |
Collapse
|
17
|
Peckham H, Webb K, Rosser EC, Butler G, Ciurtin C. Gender-Diverse Inclusion in Immunological Research: Benefits to Science and Health. Front Med (Lausanne) 2022; 9:909789. [PMID: 35911383 PMCID: PMC9329564 DOI: 10.3389/fmed.2022.909789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/24/2022] [Indexed: 01/26/2023] Open
Abstract
The differences between male and female immune systems are an under-researched field, ripe for discovery. This is evidenced by the stark sex biases seen in autoimmunity and infectious disease. Both the sex hormones (oestrogen and testosterone), as well as the sex chromosomes have been demonstrated to impact immune responses, in multiple ways. Historical shortcomings in reporting basic and clinical scientific findings in a sex-disaggregated manner have led not only to limited discovery of disease aetiology, but to potential inaccuracies in the estimation of the effects of diseases or interventions on females and gender-diverse groups. Here we propose not only that research subjects should include both cis-gender men and cis-gender women, but also transgender and gender-diverse people alongside them. The known interaction between the hormonal milieu and the sex chromosomes is inseparable in cis-gender human research, without the confounders of puberty and age. By inclusion of those pursuing hormonal affirmation of their gender identity- the individual and interactive investigation of hormones and chromosomes is permitted. Not only does this allow for a fine-tuned dissection of these individual effects, but it allows for discovery that is both pertinent and relevant to a far wider portion of the population. There is an unmet need for detailed treatment follow-up of the transgender community- little is known of the potential benefits and risks of hormonal supplementation on the immune system, nor indeed on many other health and disease outcomes. Our research team has pioneered the inclusion of gender-diverse persons in our basic research in adolescent autoimmune rheumatic diseases. We review here the many avenues that remain unexplored, and suggest ways in which other groups and teams can broaden their horizons and invest in a future for medicine that is both fruitful and inclusive.
Collapse
Affiliation(s)
- Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London, United Kingdom
- Division of Medicine, Centre for Rheumatology Research, University College London (UCL), London, United Kingdom
| | - Kate Webb
- Department of Paediatric Rheumatology, School of Child and Adolescent Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
- Crick African Network, The Francis Crick Institute, London, United Kingdom
| | - Elizabeth C. Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London, United Kingdom
- Division of Medicine, Centre for Rheumatology Research, University College London (UCL), London, United Kingdom
| | - Gary Butler
- Department of Paediatric and Adolescent Endocrinology, University College London Hospital (UCLH) and Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Gender Identity Development Service (GIDS), Tavistock and Portman NHS Foundation Trust, London, United Kingdom
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London, United Kingdom
- Division of Medicine, Centre for Rheumatology Research, University College London (UCL), London, United Kingdom
| |
Collapse
|
18
|
Activated-memory T cells influence naïve T cell fate: a noncytotoxic function of human CD8 T cells. Commun Biol 2022; 5:634. [PMID: 35768564 PMCID: PMC9243096 DOI: 10.1038/s42003-022-03596-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 06/15/2022] [Indexed: 01/26/2023] Open
Abstract
T cells are endowed with the capacity to sense their environment including other T cells around them. They do so to set their numbers and activation thresholds. This form of regulation has been well-studied within a given T cell population - i.e., within the naïve or memory pool; however, less is known about the cross-talk between T cell subsets. Here, we tested whether memory T cells interact with and influence surrounding naïve T cells. We report that human naïve CD8 T cells (TN) undergo phenotypic and transcriptional changes in the presence of autologous activated-memory CD8 T cells (TMem). Following in vitro co-culture with activated central memory cells (TCM), ~3% of the TN acquired activation/memory canonical markers (CD45RO and CD95) in an MHC-I dependent-fashion. Using scRNA-seq, we also observed that ~3% of the TN acquired an activated/memory signature, while ~84% developed a unique activated transcriptional profile hybrid between naïve and activated memory. Pseudotime trajectory analysis provided further evidence that TN with an activated/memory or hybrid phenotype were derived from TN. Our data reveal a non-cytotoxic function of TMem with potential to activate autologous TN into the activated/memory pool. These findings may have implications for host-protection and autoimmunity that arises after vaccination, infection or transplantation.
Collapse
|
19
|
Aubin AM, Lombard-Vadnais F, Collin R, Aliesky HA, McLachlan SM, Lesage S. The NOD Mouse Beyond Autoimmune Diabetes. Front Immunol 2022; 13:874769. [PMID: 35572553 PMCID: PMC9102607 DOI: 10.3389/fimmu.2022.874769] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/21/2022] [Indexed: 12/19/2022] Open
Abstract
Autoimmune diabetes arises spontaneously in Non-Obese Diabetic (NOD) mice, and the pathophysiology of this disease shares many similarities with human type 1 diabetes. Since its generation in 1980, the NOD mouse, derived from the Cataract Shinogi strain, has represented the gold standard of spontaneous disease models, allowing to investigate autoimmune diabetes disease progression and susceptibility traits, as well as to test a wide array of potential treatments and therapies. Beyond autoimmune diabetes, NOD mice also exhibit polyautoimmunity, presenting with a low incidence of autoimmune thyroiditis and Sjögren's syndrome. Genetic manipulation of the NOD strain has led to the generation of new mouse models facilitating the study of these and other autoimmune pathologies. For instance, following deletion of specific genes or via insertion of resistance alleles at genetic loci, NOD mice can become fully resistant to autoimmune diabetes; yet the newly generated diabetes-resistant NOD strains often show a high incidence of other autoimmune diseases. This suggests that the NOD genetic background is highly autoimmune-prone and that genetic manipulations can shift the autoimmune response from the pancreas to other organs. Overall, multiple NOD variant strains have become invaluable tools for understanding the pathophysiology of and for dissecting the genetic susceptibility of organ-specific autoimmune diseases. An interesting commonality to all autoimmune diseases developing in variant strains of the NOD mice is the presence of autoantibodies. This review will present the NOD mouse as a model for studying autoimmune diseases beyond autoimmune diabetes.
Collapse
Affiliation(s)
- Anne-Marie Aubin
- Immunology-Oncology Division, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Félix Lombard-Vadnais
- Immunology-Oncology Division, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Roxanne Collin
- Immunology-Oncology Division, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
- CellCarta, Montreal, QC, Canada
| | - Holly A. Aliesky
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Sandra M. McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Sylvie Lesage
- Immunology-Oncology Division, Maisonneuve-Rosemont Hospital Research Center, Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
20
|
Zhu J, Wu J, Liang Z, Mo C, Qi T, Liang S, Lian T, Qiu R, Yu X, Tang X, Wu B. Interactions between the breast tissue microbiota and host gene regulation in nonpuerperal mastitis. Microbes Infect 2022; 24:104904. [PMID: 35331909 DOI: 10.1016/j.micinf.2021.104904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/08/2021] [Accepted: 10/17/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Nonpuerperal mastitis (NPM) causes considerable psychological distress in females, since it is difficult to diagnose and treat. A spectrum of etiological factors can lead to NPM. However, the pathogenesis of NPM remains unclear. Here, we aimed to dissect the role of host gene-microbe interactions in NPM. METHODS We compared the breast tissue microbiome between NPM patients and controls using 16S rRNA sequencing. We also compared the gut microbiome between NPM patients and healthy controls. Moreover, we investigated whether the breast tissue microbiome was associated with an altered gut microbiome in patients with NPM. We analyzed differentially expressed genes in inflammatory tissues of mammary gland from patients with NPM and normal mammary gland tissues from patients with benign and non-infectious breast disease by RNA-sequencing (RNA-seq). Lastly, we explored the association of specific bacterial taxa with differential expression of immune-related genes and differences in infiltrating immune cells. RESULTS The breast tissue microbiome from NPM and controls showed significant differences in community composition. The breast tissue shared a relatively small proportion of bacterial communities with the gut in patients with NPM. Ruminococcus (family Ruminococcaceae) of breast tissue was positively correlated with the differentially expression of immune-related genes between NPM patients and controls, including antigen processing and presentation genes (ICAM1, LGMN, THBS1, TAP1, HSPA1B and HSPA1A), cytokine receptor gene IL15RA, and chemokine gene CCN1. Rhizobium of breast tissue was negatively correlated with the differentially expression of the antigen processing and presentation gene HSPA6 between NPM patients and controls. We also found that Ruminococcus (family Ruminococcaceae), Coprococcus, and Clostridium of breast tissue positively correlated with the difference of CD8+ T cells between NPM patients and controls. CONCLUSIONS We preliminarily explored the potential role of host-microbe interactions in NPM. We demonstrate cross-talk between the breast tissue microbiome and the gut microbiome in patients with NPM. We suggest that NPM microbiome composition influences the immune microenvironment of the disease by affecting the transcriptome. This is an exploratory study and further investigation of host-microbe interactions and its potential mechanism in NPM development are warranted.
Collapse
Affiliation(s)
- Jia Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Jie Wu
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Zhongzeng Liang
- Department of Breast Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, Guangdong, China
| | - Changgan Mo
- Hechi Hospital Affiliated to Youjiang Medical University for Nationalities, Hechi, 547000, Guangxi, China; The First Affiliated Hospital of Jinan University, Guangzhou, 510000, Guangdong, China
| | - Tiantian Qi
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Siyuan Liang
- Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Tao Lian
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Rongbin Qiu
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Xiaoting Yu
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Xiuge Tang
- Hechi Hospital Affiliated to Youjiang Medical University for Nationalities, Hechi, 547000, Guangxi, China.
| | - Biao Wu
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China.
| |
Collapse
|
21
|
Reitsema RD, Boots AMH, van der Geest KSM, Sandovici M, Heeringa P, Brouwer E. CD8+ T Cells in GCA and GPA: Bystanders or Active Contributors? Front Immunol 2021; 12:654109. [PMID: 33815414 PMCID: PMC8015776 DOI: 10.3389/fimmu.2021.654109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/01/2021] [Indexed: 12/28/2022] Open
Abstract
Vasculitis refers to inflammation of blood vessels and can cause a variety of serious complications depending on which vessels are affected. Two different forms of vasculitis are Giant Cell Arteritis (GCA) and Granulomatosis with Polyangiitis (GPA). GCA is the most common form of vasculitis in adults affecting the large arteries and can lead to visual impairment and development of aneurysms. GPA affects small- and medium-sized blood vessels predominantly in the lungs and kidneys resulting in organ failure. Both diseases can potentially be fatal. Although the pathogenesis of GCA and GPA are incompletely understood, a prominent role for CD4+ T cells has been implicated in both diseases. More recently, the role of CD8+ T cells has gained renewed interest. CD8+ T cells are important players in the adaptive immune response against intracellular microorganisms. After a general introduction on the different forms of vasculitis and their association with infections and CD8+ T cells, we review the current knowledge on CD8+ T-cell involvement in the immunopathogenesis of GCA and GPA focusing on phenotypic and functional features of circulating and lesional CD8+ T cells. Furthermore, we discuss to which extent aging is associated with CD8+ T-cell phenotype and function in GCA and GPA.
Collapse
Affiliation(s)
- Rosanne D Reitsema
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Annemieke M H Boots
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Kornelis S M van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
22
|
Wu H, Estrella V, Beatty M, Abrahams D, El-Kenawi A, Russell S, Ibrahim-Hashim A, Longo DL, Reshetnyak YK, Moshnikova A, Andreev OA, Luddy K, Damaghi M, Kodumudi K, Pillai SR, Enriquez-Navas P, Pilon-Thomas S, Swietach P, Gillies RJ. T-cells produce acidic niches in lymph nodes to suppress their own effector functions. Nat Commun 2020; 11:4113. [PMID: 32807791 PMCID: PMC7431837 DOI: 10.1038/s41467-020-17756-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 07/13/2020] [Indexed: 11/27/2022] Open
Abstract
The acidic pH of tumors profoundly inhibits effector functions of activated CD8 + T-cells. We hypothesize that this is a physiological process in immune regulation, and that it occurs within lymph nodes (LNs), which are likely acidic because of low convective flow and high glucose metabolism. Here we show by in vivo fluorescence and MR imaging, that LN paracortical zones are profoundly acidic. These acidic niches are absent in athymic Nu/Nu and lymphodepleted mice, implicating T-cells in the acidifying process. T-cell glycolysis is inhibited at the low pH observed in LNs. We show that this is due to acid inhibition of monocarboxylate transporters (MCTs), resulting in a negative feedback on glycolytic rate. Importantly, we demonstrate that this acid pH does not hinder initial activation of naïve T-cells by dendritic cells. Thus, we describe an acidic niche within the immune system, and demonstrate its physiological role in regulating T-cell activation.
Collapse
Affiliation(s)
- Hao Wu
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
- Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, P.R. China
| | - Veronica Estrella
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Matthew Beatty
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Dominique Abrahams
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Asmaa El-Kenawi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Shonagh Russell
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Arig Ibrahim-Hashim
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Turin, Italy
| | - Yana K Reshetnyak
- Department of Physics, University of Rhode Island, Kingston, RI, 02881, USA
| | - Anna Moshnikova
- Department of Physics, University of Rhode Island, Kingston, RI, 02881, USA
| | - Oleg A Andreev
- Department of Physics, University of Rhode Island, Kingston, RI, 02881, USA
| | - Kimberly Luddy
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Mehdi Damaghi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Krithika Kodumudi
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Smitha R Pillai
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Pedro Enriquez-Navas
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, England, UK.
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
23
|
El-Zaatari M, Bishu S, Zhang M, Grasberger H, Hou G, Haley H, Humphries B, Syu LJ, Dlugosz AA, Luker K, Luker GD, Eaton K, Kamada N, Cascalho M, Kao JY. Aim2-mediated/IFN-β-independent regulation of gastric metaplastic lesions via CD8+ T cells. JCI Insight 2020; 5:e94035. [PMID: 32053518 PMCID: PMC7141403 DOI: 10.1172/jci.insight.94035] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 02/12/2020] [Indexed: 01/24/2023] Open
Abstract
Development of gastric cancer is often preceded by chronic inflammation, but the immune cellular mechanisms underlying this process are unclear. Here we demonstrated that an inflammasome molecule, absent in melanoma 2 (Aim2), was upregulated in patients with gastric cancer and in spasmolytic polypeptide-expressing metaplasia of chronically Helicobacter felis-infected stomachs in mice. However, we found that Aim2 was not necessary for inflammasome function during gastritis. In contrast, Aim2 deficiency led to an increase in gastric CD8+ T cell frequency, which exacerbated metaplasia. These gastric CD8+ T cells from Aim2-/- mice were found to have lost their homing receptor expression (sphingosine-1-phosphate receptor 1 [S1PR1] and CD62L), a feature of tissue-resident memory T cells. The process was not mediated by Aim2-dependent regulation of IFN-β or by dendritic cell-intrinsic Aim2. Rather, Aim2 deficiency contributed to an increased production of CXCL16 by B cells, which could suppress S1PR1 and CD62L in CD8+ T cells. This study describes a potentially novel function of Aim2 that regulates CD8+ T cell infiltration and retention within chronically inflamed solid organ tissue. This function operates independent of the inflammasome, IFN-β, or dendritic cells. We provide evidence that B cells can contribute to this mechanism via CXCL16.
Collapse
Affiliation(s)
- Mohamad El-Zaatari
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Shrinivas Bishu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Min Zhang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Helmut Grasberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Guoqing Hou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Henry Haley
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Brock Humphries
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Li-Jyun Syu
- Department of Dermatology, School of Medicine
| | | | - Kathy Luker
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Gary D. Luker
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | | | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Marilia Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - John Y. Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| |
Collapse
|
24
|
Yang H, Yao Z, Zhou X, Zhang W, Zhang X, Zhang F. Immune-related adverse events of checkpoint inhibitors: Insights into immunological dysregulation. Clin Immunol 2020; 213:108377. [PMID: 32135278 DOI: 10.1016/j.clim.2020.108377] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/01/2020] [Accepted: 03/01/2020] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) targeting against programmed cell death-1(PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) have shown efficacy in cancer treatment. However, a spectrum of immune-related adverse events (irAEs) have raised concerns about their clinical application. IrAEs are distinct from traditional chemo- and radiotherapy-induced toxicities, as they are related in particular to the dysregulation of immune system and autoimmunity. The underlying pathogenesis of irAEs remains elusive. Understanding of the potential underlying mechanism is of great importance for the management of irAEs and the development of new ICIs with insignificant irAEs. In this review, we summarize the current evidence to provide insights into the biological basis of irAEs and provide a potential explanation for their pathogenesis, with focus on the relationship between checkpoint molecules and immune cell regulation.
Collapse
Affiliation(s)
- Huaxia Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing 100730, China; Clinical Immunology Center, Medical Epigenetics Research Center, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Zhuoran Yao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing 100730, China
| | - Xiaoxiang Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing 100730, China
| | - Wen Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing 100730, China; Clinical Immunology Center, Medical Epigenetics Research Center, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing 100730, China; Clinical Immunology Center, Medical Epigenetics Research Center, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China.
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing 100730, China; Clinical Immunology Center, Medical Epigenetics Research Center, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China.
| |
Collapse
|
25
|
Kim KH, Kim CG, Shin EC. Peripheral blood immune cell-based biomarkers in anti-PD-1/PD-L1 therapy. Immune Netw 2020; 20:e8. [PMID: 32158596 PMCID: PMC7049582 DOI: 10.4110/in.2020.20.e8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint blockade targeting PD-1 and PD-L1 has resulted in unprecedented clinical benefit for cancer patients. Anti-PD-1/PD-L1 therapy has become the standard treatment for diverse cancer types as monotherapy or in combination with other anti-cancer therapies, and its indications are expanding. However, many patients do not benefit from anti-PD-1/PD-L1 therapy due to primary and/or acquired resistance, which is a major obstacle to broadening the clinical applicability of anti-PD-1/PD-L1 therapy. In addition, hyperprogressive disease, an acceleration of tumor growth following anti-PD-1/PD-L1 therapy, has been proposed as a new response pattern associated with deleterious prognosis. Anti-PD-1/PD-L1 therapy can also cause a unique pattern of adverse events termed immune-related adverse events, sometimes leading to treatment discontinuation and fatal outcomes. Investigations have been carried out to predict and monitor treatment outcomes using peripheral blood as an alternative to tissue biopsy. This review summarizes recent studies utilizing peripheral blood immune cells to predict various outcomes in cancer patients treated with anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Kyung Hwan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Chang Gon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
26
|
Kim KH, Hur JY, Cho J, Ku BM, Koh J, Koh JY, Sun JM, Lee SH, Ahn JS, Park K, Ahn MJ, Shin EC. Immune-related adverse events are clustered into distinct subtypes by T-cell profiling before and early after anti-PD-1 treatment. Oncoimmunology 2020; 9:1722023. [PMID: 32076579 PMCID: PMC6999841 DOI: 10.1080/2162402x.2020.1722023] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/17/2019] [Accepted: 11/04/2019] [Indexed: 12/31/2022] Open
Abstract
Although anti-programmed death-1 (PD-1) treatment has shown remarkable anti-tumor efficacy, immune-related adverse events (irAEs) develop with heterogeneous clinical manifestations. However, the immunological understanding of irAEs is currently limited. In the present study, we analyzed peripheral blood T cells obtained from cancer patients who received anti-PD-1 treatment to determine the immunological characteristics of irAEs. This study included 31 patients with refractory thymic epithelial tumor (TET) who were enrolled in a phase II trial of pembrolizumab (NCT02607631) and 60 patients with metastatic non-small cell lung cancer (NSCLC) who received pembrolizumab or nivolumab. T-cell profiling was performed by multicolor flow cytometry using peripheral blood obtained before treatment and 7 days after the first dose of anti-PD-1 antibodies. irAEs developed in 21 TET patients and 24 NSCLC patients. Severe (≥ grade 3) irAEs occurred in 7 TET patients (22.6%) and 6 NSCLC patients (10.0%). Patients with severe irAEs exhibited a significantly lower fold increase in the frequency of effector regulatory T (eTreg) cells after anti-PD-1 treatment, a higher ratio of T helper-17 (Th17) and T helper-1 cells at baseline, and a higher percentage of Ki-67+ cells among PD-1+CD8+ T cells posttreatment. In clustering analysis using the T-cell parameters, patients with irAEs were grouped into four distinct subtypes: Th17-related, TNF-related, CD8-related Treg-compensated, and CD8-related Treg-uncompensated. The T-cell parameters showed a predictive value for the development of each subtype of severe irAEs. In conclusion, severe irAEs after anti-PD-1 treatment were clustered into four immunological subtypes, and potential biomarkers for early prediction of severe irAEs were proposed.
Collapse
Affiliation(s)
- Kyung Hwan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Joon Young Hur
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jinhyun Cho
- Division of Hematology-Oncology, Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Republic of Korea
| | - Bo Mi Ku
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jiae Koh
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - June Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
27
|
Kiaee F, Azizi G, Rafiemanesh H, Zainaldain H, Sadaat Rizvi F, Alizadeh M, Jamee M, Mohammadi S, Habibi S, Sharifi L, Jadidi-Niaragh F, Haghi S, Yazdani R, Abolhassani H, Aghamohammadi A. Malignancy in common variable immunodeficiency: a systematic review and meta-analysis. Expert Rev Clin Immunol 2019; 15:1105-1113. [PMID: 31452405 DOI: 10.1080/1744666x.2019.1658523] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background: Common variable immunodeficiency (CVID) is the most common clinically significant primary immunodeficiency (PID) disorder characterized by variable clinical manifestations including recurrent infections, autoimmune disorders, enteropathy, lymphoproliferative disorders, and malignancy. The aim of this study is to estimate the overall prevalence of malignancy in patients with CVID. Methods: PubMed, Web of Science and Scopus were searched systemically to find eligible studies from the earliest available date to March 2019 with standard keywords. Pooled estimates of the malignancy prevalence and the corresponding 95% confidence intervals (CI) were calculated using random effects models. Results: Forty-eight studies with a total of 8123 CVID patients met the inclusion criteria and were finally included in the meta-analysis. Overall prevalence of malignancy was 8.6% (95% CI: 7.1-10.0; I2 = 79.2%). The prevalence of lymphoma, gastric cancer, and breast cancer in CVID patients were 4.1% (95% CI: 3.3-4.9; I2 = 62.6%), 1.5% (95% CI: 0.78-2.2; I2 = 68.9%), and 1.3% (95% CI: 0.64-1.9; I2 = 54.9%), respectively. Moreover, autoimmunity and malabsorption were more frequent in patients with malignancy than those without malignancy. Conclusion: The prevalence of malignancy has increased in CVID patients due to recent improvement in survival rate and the lymphoma is the most common type. This research highlighted the significance of malignancy screening and management in CVID patients.
Collapse
Affiliation(s)
- Fatemeh Kiaee
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences , Tehran , Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences , Karaj , Iran
| | - Hosein Rafiemanesh
- Student Research Committee, Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Hamed Zainaldain
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Fatema Sadaat Rizvi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Mahla Alizadeh
- Evidence- Based Phytotherapy and Complementary Medicine Research Center, Alborz University of Medical Sciences , Karaj , Iran.,Student Research Committee, Alborz University of Medical Sciences , Karaj , Iran
| | - Mahnaz Jamee
- Student Research Committee, Alborz University of Medical Sciences , Karaj , Iran
| | - Sara Mohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Sima Habibi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Laleh Sharifi
- Uro-Oncology Research Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Sabahat Haghi
- Department of Hematology & Oncology, School of Medicine, Alborz University of Medical Sciences , Karaj , Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge , Stockholm , Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
28
|
Dong MB, Wang G, Chow RD, Ye L, Zhu L, Dai X, Park JJ, Kim HR, Errami Y, Guzman CD, Zhou X, Chen KY, Renauer PA, Du Y, Shen J, Lam SZ, Zhou JJ, Lannin DR, Herbst RS, Chen S. Systematic Immunotherapy Target Discovery Using Genome-Scale In Vivo CRISPR Screens in CD8 T Cells. Cell 2019; 178:1189-1204.e23. [PMID: 31442407 PMCID: PMC6719679 DOI: 10.1016/j.cell.2019.07.044] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/17/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022]
Abstract
CD8 T cells play essential roles in anti-tumor immune responses. Here, we performed genome-scale CRISPR screens in CD8 T cells directly under cancer immunotherapy settings and identified regulators of tumor infiltration and degranulation. The in vivo screen robustly re-identified canonical immunotherapy targets such as PD-1 and Tim-3, along with genes that have not been characterized in T cells. The infiltration and degranulation screens converged on an RNA helicase Dhx37. Dhx37 knockout enhanced the efficacy of antigen-specific CD8 T cells against triple-negative breast cancer in vivo. Immunological characterization in mouse and human CD8 T cells revealed that DHX37 suppresses effector functions, cytokine production, and T cell activation. Transcriptomic profiling and biochemical interrogation revealed a role for DHX37 in modulating NF-κB. These data demonstrate high-throughput in vivo genetic screens for immunotherapy target discovery and establishes DHX37 as a functional regulator of CD8 T cells.
Collapse
Affiliation(s)
- Matthew B Dong
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Yale MD-PhD Program, Yale University School of Medicine, New Haven, CT 06510, USA; Immunobiology Program, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Guangchuan Wang
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA
| | - Ryan D Chow
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Yale MD-PhD Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Lupeng Ye
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA
| | - Lvyun Zhu
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA
| | - Xiaoyun Dai
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA
| | - Jonathan J Park
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Yale MD-PhD Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Hyunu R Kim
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA
| | - Youssef Errami
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA
| | - Christopher D Guzman
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Immunobiology Program, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Combined Program in the Biological and Biomedical Sciences, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xiaoyu Zhou
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA
| | - Krista Y Chen
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; The College, Yale University, New Haven, CT 06520, USA
| | - Paul A Renauer
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Combined Program in the Biological and Biomedical Sciences, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yaying Du
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA
| | - Johanna Shen
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; The College, Yale University, New Haven, CT 06520, USA
| | - Stanley Z Lam
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; The College, Yale University, New Haven, CT 06520, USA
| | - Jingjia J Zhou
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; The College, Yale University, New Haven, CT 06520, USA
| | - Donald R Lannin
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06510, USA; Breast Cancer Program, Yale University School of Medicine, New Haven, CT06510, USA; Smilow Cancer Hospital, 35 Park Street, New Haven, CT 06510; Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Roy S Herbst
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA; Smilow Cancer Hospital, 35 Park Street, New Haven, CT 06510; Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; System Biology Institute, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Center for Cancer Systems Biology, Integrated Science & Technology Center, Yale University, 850 W Campus Drive, West Haven, CT 06516, USA; Yale MD-PhD Program, Yale University School of Medicine, New Haven, CT 06510, USA; Immunobiology Program, Yale University School of Medicine, New Haven, CT 06510, USA; Combined Program in the Biological and Biomedical Sciences, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Liver Center, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
29
|
Suppression of a broad spectrum of liver autoimmune pathologies by single peptide-MHC-based nanomedicines. Nat Commun 2019; 10:2150. [PMID: 31089130 PMCID: PMC6517389 DOI: 10.1038/s41467-019-09893-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/05/2019] [Indexed: 01/28/2023] Open
Abstract
Peptide-major histocompatibility complex class II (pMHCII)-based nanomedicines displaying tissue-specific autoantigenic epitopes can blunt specific autoimmune conditions by re-programming cognate antigen-experienced CD4+ T-cells into disease-suppressing T-regulatory type 1 (TR1) cells. Here, we show that single pMHCII-based nanomedicines displaying epitopes from mitochondrial, endoplasmic reticulum or cytoplasmic antigens associated with primary biliary cholangitis (PBC) or autoimmune hepatitis (AIH) can broadly blunt PBC, AIH and Primary Sclerosing Cholangitis in various murine models in an organ- rather than disease-specific manner, without suppressing general or local immunity against infection or metastatic tumors. Therapeutic activity is associated with cognate TR1 cell formation and expansion, TR1 cell recruitment to the liver and draining lymph nodes, local B-regulatory cell formation and profound suppression of the pro-inflammatory capacity of liver and liver-proximal myeloid dendritic cells and Kupffer cells. Thus, autoreactivity against liver-enriched autoantigens in liver autoimmunity is not disease-specific and can be harnessed to treat various liver autoimmune diseases broadly. Immune response against tissue-specific antigens is a hallmark of autoimmunity. Here the authors show that a single autoantigen-based nanomedicine can ameliorate pathology in a broad range of liver autoimmunity models without impairing host defenses, suggesting organ-wide tolerization.
Collapse
|
30
|
Deng Q, Luo Y, Chang C, Wu H, Ding Y, Xiao R. The Emerging Epigenetic Role of CD8+T Cells in Autoimmune Diseases: A Systematic Review. Front Immunol 2019; 10:856. [PMID: 31057561 PMCID: PMC6482221 DOI: 10.3389/fimmu.2019.00856] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
Autoimmune diseases are usually complex and multifactorial, characterized by aberrant production of autoreactive immune cells and/or autoantibodies against healthy cells and tissues. However, the pathogenesis of autoimmune diseases has not been clearly elucidated. The activation, differentiation, and development of CD8+ T cells can be affected by numerous inflammatory cytokines, transcription factors, and chemokines. In recent years, epigenetic modifications have been shown to play an important role in the fate of CD8+ T cells. The discovery of these modifications that contribute to the activation or suppression of CD8+ cells has been concurrent with the increasing evidence that CD8+ T cells play a role in autoimmunity. These relationships have been studied in various autoimmune diseases, including multiple sclerosis (MS), systemic sclerosis (SSc), type 1 diabetes (T1D), Grave's disease (GD), systemic lupus erythematosus (SLE), aplastic anemia (AA), and vitiligo. In each of these diseases, genes that play a role in the proliferation or activation of CD8+ T cells have been found to be affected by epigenetic modifications. Various cytokines, transcription factors, and other regulatory molecules have been found to be differentially methylated in CD8+ T cells in autoimmune diseases. These genes are involved in T cell regulation, including interferons, interleukin (IL),tumor necrosis factor (TNF), as well as linker for activation of T cells (LAT), cytotoxic T-lymphocyte–associated antigen 4 (CTLA4), and adapter proteins. MiRNAs also play a role in the pathogenesis of these diseases and several known miRNAs that are involved in these diseases have also been shown to play a role in CD8+ regulation.
Collapse
Affiliation(s)
- Qiancheng Deng
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yangyang Luo
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Children's Hospital, Changsha, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Ding
- Department of Dermatology, Hainan Provincial Dermatology Disease Hospital, Haikou, China
| | - Rong Xiao
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
31
|
Chen S, Pu W, Guo S, Jin L, He D, Wang J. Genome-Wide DNA Methylation Profiles Reveal Common Epigenetic Patterns of Interferon-Related Genes in Multiple Autoimmune Diseases. Front Genet 2019; 10:223. [PMID: 31024609 PMCID: PMC6459983 DOI: 10.3389/fgene.2019.00223] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 02/28/2019] [Indexed: 11/13/2022] Open
Abstract
Graves' disease (GD), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) are complex autoimmune diseases sharing common clinical, genetic and pathogenetic features. However, the commonalities of the DNA methylation profiles for these diseases are still unknown. We conducted an integrative analysis of the multiple-autoimmune disease methylation dataset including GD, RA, SLE, and SSc samples, to identify the common methylation patterns of autoimmune diseases. We identified 15,289 differentially methylated sites between multiple-autoimmune disease patients and controls in CD4+ T cells. We found that the most significant differentially methylated sites had a remarkable enrichment in type I interferon (IFN) pathway genes. Similarly, we identified 9,295 differentially methylated sites between GD/SSc patients and controls in CD8+ T cells. The overall IFN-related gene panel annotated by gene ontology (GO) showed an excellent diagnostic capacity in CD4+ T cells (Sensitivity = 0.82, specificity = 0.82 and AUC = 0.90), while IFI44L, another IFN-related gene not annotated by GO, showed high prediction ability in both CD4+ (AUC = 0.86) and CD8+ (AUC = 0.75) T cells. In conclusion, our study demonstrated that hypomethylation of IFN-related genes is a common feature of GD/RA/SLE/SSc patients in CD4+ T cells, and the DNA methylation profile of IFN-related genes could be promising biomarkers for the diagnosis of GD, RA, SLE, and SSc.
Collapse
Affiliation(s)
- Sidi Chen
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Weilin Pu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Shicheng Guo
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI, United States
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrated Traditional and Western Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Traditional Chinese Medicine Research Institute, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Aurora A controls CD8 + T cell cytotoxic activity and antiviral response. Sci Rep 2019; 9:2211. [PMID: 30778113 PMCID: PMC6379542 DOI: 10.1038/s41598-019-38647-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/28/2018] [Indexed: 12/05/2022] Open
Abstract
Aurora A is a serine/threonine kinase whose role in cell cycle progression and tumour generation has been widely studied. Recent work has revealed an unexpected function for Aurora A during CD4+ T cell activation and, also, in graft versus host disease development. However, it remains unknown whether Aurora A is involved in CD8+ T cell effector function and in cytotoxic T lymphocyte-mediated antiviral response. Here, we show that Aurora A chemical inhibition leads to an impairment of both the peptide-specific cytotoxicity and the degranulation activity of CD8+ T cells. This finding was similarly proven for both mice and human CD8+ CTL activity. As a result of Aurora A blockade, we detected a reduction in the expression induced by T cell activation of genes classically related to the effector function of cytotoxic T lymphocytes such as granzyme B or perforin1. Finally, we have found that Aurora A is necessary for CD8+ T cell-mediated antiviral response, in an in vivo model of vaccinia virus infection. Thus, we can conclude that Aurora A activity is, indeed, needed for the proper effector function of cytotoxic T lymphocytes and for their activity against viral threats.
Collapse
|
33
|
Harms RZ, Lorenzo-Arteaga KM, Ostlund KR, Smith VB, Smith LM, Gottlieb P, Sarvetnick N. Abnormal T Cell Frequencies, Including Cytomegalovirus-Associated Expansions, Distinguish Seroconverted Subjects at Risk for Type 1 Diabetes. Front Immunol 2018; 9:2332. [PMID: 30405601 PMCID: PMC6204396 DOI: 10.3389/fimmu.2018.02332] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/19/2018] [Indexed: 12/12/2022] Open
Abstract
We analyzed T cell subsets from cryopreserved PBMC obtained from the TrialNet Pathway to Prevention archives. We compared subjects who had previously seroconverted for one or more autoantibodies with non-seroconverted, autoantibody negative individuals. We observed a reduced frequency of MAIT cells among seroconverted subjects. Seroconverted subjects also possessed decreased frequencies of CCR4-expressing CD4 T cells, including a regulatory-like subset. Interestingly, we found an elevation of CD57+, CD28–, CD127–, CD27– CD8 T cells (SLEC) among seroconverted subjects that was most pronounced among those that progressed to disease. The frequency of these SLEC was strongly correlated with CMV IgG abundance among seroconverted subjects, associated with IA-2 levels, and most elevated among CMV+ seroconverted subjects who progressed to disease. Combined, our data indicate discrete, yet profound T cell alterations are associated with islet autoimmunity among at-risk subjects.
Collapse
Affiliation(s)
- Robert Z Harms
- Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
| | | | - Katie R Ostlund
- Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
| | - Victoria B Smith
- Office of the Vice Chancellor of Research, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lynette M Smith
- Biostatistics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Peter Gottlieb
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nora Sarvetnick
- Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
34
|
Kang N, Toyofuku WM, Yang X, Scott MD. Inhibition of allogeneic cytotoxic T cell (CD8 +) proliferation via polymer-induced Treg (CD4 +) cells. Acta Biomater 2017; 57:146-155. [PMID: 28442414 DOI: 10.1016/j.actbio.2017.04.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/13/2017] [Accepted: 04/21/2017] [Indexed: 01/16/2023]
Abstract
T cell-mediated immune rejection remains a barrier to successful transplantation. Polymer-based bioengineering of cells may provide an effective means of preventing allorecognition and the proliferation of cytotoxic (CD8+) T lymphocytes (CTL). Using MHC-disparate murine splenocytes modified with succinimidyl valerate activated methoxypoly(ethylene glycol) [SVA-mPEG] polymers, the effects of leukocyte immunocamouflage on CD8+ and CD4+ alloproliferation and T regulatory (Treg) cell induction were assessed in a mixed lymphocyte reaction (MLR) model. Polymer-grafting effectively camouflaged multiple leukocyte markers (MHC class I and II, TCR and CD3) essential for effective allorecognition. Consequent to the polymer-induced immunocamouflage of the cell membrane, both CD8+ and CD4+ T cell alloproliferation were significantly inhibited in a polymer dose-dependent manner. The loss of alloproliferation correlated with the induction of Treg cells (CD4+CD25+Foxp3+). The Tregs, surprisingly, arose primarily via differentiation of naive, non-proliferating, CD4+ cells. Of biologic importance, the polymer-induced Treg were functional and exhibited potent immunosuppressive activity on allogeneic CTL proliferation. These results suggest that immunocamouflage-mediated attenuation of alloantigen-TCR recognition can prevent the tissue destructive allogeneic CD8+ T cell response, both directly and indirectly, through the generation/differentiation of functional Tregs. Immunocamouflage induced tolerance could be clinically valuable in attenuating T cell-mediated transplant rejection and in the treatment of autoimmune diseases. STATEMENT OF SIGNIFICANCE While our previous studies have demonstrated that polymer-grafting to MHC disparate leukocytes inhibits CD4+ cell proliferation, the effects of PEGylation on the alloproliferation of CD8+ cytotoxic T cells (CTL) was not examined. As shown here, PEGylation of allogeneic leukocytes prevents the generation of the CTL response responsible for acute rejection. The loss of CTL proliferation is consequent to the polymer-based attenuation of allorecognition and the induction of T regulatory cells (Tregs). Interestingly, the Tregs are primarily generated via the differentiation of non-proliferating naive T cells. Importantly, the Tregs are functional and effectively induce a tolerogenic environment when transferred to an alloresponsive environment. The use of polymer-modified leukocytes provides a unique approach to effectively maximize the biologic production of functional Tregs both in vitro and in vivo. By using this approach it may be possible to attenuate unwanted alloresponses (e.g., graft rejection) or to treat autoimmune diseases.
Collapse
Affiliation(s)
- Ning Kang
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Wendy M Toyofuku
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Xining Yang
- University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Mark D Scott
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
35
|
Kujur W, Gurram RK, Maurya SK, Nadeem S, Chodisetti SB, Khan N, Agrewala JN. Caerulomycin A suppresses the differentiation of naïve T cells and alleviates the symptoms of experimental autoimmune encephalomyelitis. Autoimmunity 2017; 50:317-328. [PMID: 28686480 DOI: 10.1080/08916934.2017.1332185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a highly detrimental autoimmune disease of the central nervous system. There is no cure for it but the treatment typically focuses on subsiding severity and recurrence of the disease. Experimental autoimmune encephalomyelitis (EAE) is an animal model of MS. It is characterized by frequent relapses due to the generation of memory T cells. Caerulomycin A (CaeA) is known to suppress the Th1 cells, Th2 cells, and Th17 cells. Interestingly, it enhances the generation of regulatory T cells (Tregs). Th1 cells and Th17 cells are known to aggravate EAE, whereas Tregs suppress the disease symptoms. Consequently, in the current study we evaluated the influence of CaeA on EAE. Intriguingly, we observed by whole body imaging that CaeA regressed the clinical symptoms of EAE. Further, there was reduction in the pool of Th1 cells, Th17 cells, and CD8 T cells. The mechanism involved in suppressing the EAE symptoms was due to the inhibition in the generation of effector and central memory T cells and induction of the expansion of Tregs. In essence, these findings implicate that CaeA may be considered as a potent future immunosuppressive drug.
Collapse
Affiliation(s)
- Weshely Kujur
- a Immunology Laboratory , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Rama Krishna Gurram
- a Immunology Laboratory , CSIR-Institute of Microbial Technology , Chandigarh , India.,b National Institute of Allergy and Infectious Diseases , Bethesda , MD , USA
| | - Sudeep K Maurya
- a Immunology Laboratory , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Sajid Nadeem
- a Immunology Laboratory , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Sathi Babu Chodisetti
- a Immunology Laboratory , CSIR-Institute of Microbial Technology , Chandigarh , India.,c Department of Microbiology and Immunology , Pennsylvania State University College of Medicine , Hershey , PA , USA
| | - Nargis Khan
- a Immunology Laboratory , CSIR-Institute of Microbial Technology , Chandigarh , India.,d Department of Medicine , McGill University , Montreal , QC , Canada
| | - Javed Naim Agrewala
- a Immunology Laboratory , CSIR-Institute of Microbial Technology , Chandigarh , India
| |
Collapse
|
36
|
Coppola A, Tomasello L, Pitrone M, Cillino S, Richiusa P, Pizzolanti G, Giordano C. Human limbal fibroblast-like stem cells induce immune-tolerance in autoreactive T lymphocytes from female patients with Hashimoto's thyroiditis. Stem Cell Res Ther 2017; 8:154. [PMID: 28673339 PMCID: PMC5496215 DOI: 10.1186/s13287-017-0611-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/02/2017] [Accepted: 06/15/2017] [Indexed: 12/29/2022] Open
Abstract
Background Due to their “natural immune privilege” and immunoregulatory properties human fibroblast-like limbal stem cells (f-LSCs) have acquired great interest as a potential tool for achieving immunotolerance. Hashimoto’s thyroiditis (HT) is the most common thyroid autoimmune disease and cause of hypothyroidism. To date, conventional hormone replacement therapy and unspecific immunosuppressive regimens cannot provide a definitive cure for HT subjects. We explored the immunosuppressant potential of human f-LSCs on circulating lymphomonocytes (PBMCs) collected from healthy donors and female HT patients. Methods We assessed the immunophenotyping of f-LSCs, both untreated and after 48 h of proinflammatory cytokine exposure, by means of quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and flow cytometry. The immunosuppressant effects of f-LSCs on healthy activated PBMCs were investigated in cell-cell contact and transwell settings through cell cycle assay, acridine orange staining, and caspase-3 detection. We also studied T-cell responses and possible Treg conversion by means of flow cytometry. Functional assays were conducted in activated HT lymphocytes cocultured with f-LSCs after carboxyfluorescein succinimidyl ester labeling and intracellular detection of pro- and anti-inflammatory cytokines. Results The hypo-immunogenicity of the f-LSC population depended on both cell contact and soluble factors produced, as well as the undetectable expression of all those molecules required to fully activate T lymphocytes. Following exposure to Th1 cytokines, f-LSCs augmented expression of programmed death-ligand 1 and 2 (PDL-1 and -2), indoleamine-pyrrole-2,3-dioxygenase (IDO), interleukin (IL)-6, and monocyte chemotactic protein 1 (MCP-1) while maintaining their negative phenotype for major histocompatibility (MHC) class II and costimulatory molecules. During coculture, f-LSCs suppressed up to 40% of proliferation in healthy activated PBMCs, arrested them in the G0/G1 cell cycle phase without inducing apoptosis cascade, inverted the CD4/CD8 ratio, and promoted sustained expression of the immunomodulator marker CD69. Under coculture conditions the Th imbalance of autoreactive T cells from female HT patients was fully restored. Conclusions Our study describes an in vitro coculture system able to prevent inappropriate activation of autoreactive T lymphocytes of female HT patients and to generate a tolerogenic environment even in an inflammatory background. Further investigations are necessary to establish whether this stem cell-based therapy approach in HT could avoid lifetime hormone replacement therapy by inducing T-cell education. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0611-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Antonina Coppola
- Laboratory of Regenerative Medicine, Section of Endocrinology, Diabetology and Metabolism, Di.Bi.M.I.S., University of Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy.,ATeN (Advanced Technologies Network Center), University of Palermo, Palermo, Italy
| | - Laura Tomasello
- Laboratory of Regenerative Medicine, Section of Endocrinology, Diabetology and Metabolism, Di.Bi.M.I.S., University of Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy.,ATeN (Advanced Technologies Network Center), University of Palermo, Palermo, Italy
| | - Maria Pitrone
- Laboratory of Regenerative Medicine, Section of Endocrinology, Diabetology and Metabolism, Di.Bi.M.I.S., University of Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy.,ATeN (Advanced Technologies Network Center), University of Palermo, Palermo, Italy
| | | | - Pierina Richiusa
- Laboratory of Regenerative Medicine, Section of Endocrinology, Diabetology and Metabolism, Di.Bi.M.I.S., University of Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy
| | - Giuseppe Pizzolanti
- Laboratory of Regenerative Medicine, Section of Endocrinology, Diabetology and Metabolism, Di.Bi.M.I.S., University of Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy. .,ATeN (Advanced Technologies Network Center), University of Palermo, Palermo, Italy.
| | - Carla Giordano
- Laboratory of Regenerative Medicine, Section of Endocrinology, Diabetology and Metabolism, Di.Bi.M.I.S., University of Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy. .,ATeN (Advanced Technologies Network Center), University of Palermo, Palermo, Italy.
| |
Collapse
|
37
|
High levels of soluble GPR56/ADGRG1 are associated with positive rheumatoid factor and elevated tumor necrosis factor in patients with rheumatoid arthritis. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2017; 51:485-491. [PMID: 28690029 DOI: 10.1016/j.jmii.2016.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/26/2016] [Accepted: 11/07/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND GPR56/ADGRG1 is a member of the adhesion-class G protein-coupled receptor (aGPCR) family important in brain development, oncogenesis and tumor metastasis. Like other aGPCRs, GPR56 is cleaved at the GPCR proteolysis site (GPS) motif into an N-terminal fragment (NTF) and a C-terminal fragment (CTF). Existence of soluble GPR56 (sGPR56) has been shown in vitro, however the underlying mechanism and its pathophysiologic role remains undetermined. OBJECTIVE To assess the presence of sGPR56 in human serum using ELISA assay and compare the serum sGPR56 levels among patients of various chronic inflammatory diseases and healthy subjects. PATIENTS AND METHODS In this study, serum samples from patients with systemic lupus erythematosus (SLE) (n = 57), rheumatoid arthritis (RA) (n = 95), Sjögren's syndrome (SS) (n = 29), ankylosing spondylitis (AS) (n = 51), and normal controls (n = 81) were analyzed using sGPR56-specific ELISA. RESULT We show that serum sGPR56 levels are increased in patients of RA, but not in those with SLE, SS and AS. Intriguingly, serum sGPR56 levels in RA patients correlated with positive rheumatoid factor, a marker of bone erosion and poor outcome. In addition, an elevated sGPR56 level is also noted in RA patients with higher tumor necrosis factor level. CONCLUSION we conclude that sGPR56 is present in vivo and sGPR56 level is elevated in certain chronic inflammatory diseases such as RA. Hence, sGPR56 might be considered a potential biomarker for RA disease progression.
Collapse
|
38
|
Kyaw T, Peter K, Li Y, Tipping P, Toh BH, Bobik A. Cytotoxic lymphocytes and atherosclerosis: significance, mechanisms and therapeutic challenges. Br J Pharmacol 2017; 174:3956-3972. [PMID: 28471481 DOI: 10.1111/bph.13845] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 04/02/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023] Open
Abstract
Cytotoxic lymphocytes encompass natural killer lymphocytes (cells) and cytotoxic T cells that include CD8+ T cells, natural killer (NK) T cells, γ, δ (γδ)-T cells and human CD4 + CD28- T cells. These cells play critical roles in inflammatory diseases and in controlling cancers and infections. Cytotoxic lymphocytes can be activated via a number of mechanisms that may involve dendritic cells, macrophages, cytokines or surface proteins on stressed cells. Upon activation, they secrete pro-inflammatory cytokines as well as anti-inflammatory cytokines, chemokines and cytotoxins to promote inflammation and the development of atherosclerotic lesions including vulnerable lesions, which are strongly implicated in myocardial infarctions and strokes. Here, we review the mechanisms that activate and regulate cytotoxic lymphocyte activity, including activating and inhibitory receptors, cytokines, chemokine receptors-chemokine systems utilized to home to inflamed lesions and cytotoxins and cytokines through which they affect other cells within lesions. We also examine their roles in human and mouse models of atherosclerosis and the mechanisms by which they exert their pathogenic effects. Finally, we discuss strategies for therapeutically targeting these cells to prevent the development of atherosclerotic lesions and vulnerable plaques and the challenge of developing highly targeted therapies that only minimally affect the body's immune system, avoiding the complications, such as increased susceptibility to infections, which are currently associated with many immunotherapies for autoimmune diseases. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Tin Kyaw
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Immunology, Monash University, Melbourne, Vic, Australia
| | - Yi Li
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Peter Tipping
- Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Ban-Hock Toh
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| | - Alex Bobik
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.,Department of Immunology, Monash University, Melbourne, Vic, Australia.,Department of Medicine, Monash University, Melbourne, Vic, Australia
| |
Collapse
|
39
|
Barr JY, Wang X, Meyerholz DK, Lieberman SM. CD8 T cells contribute to lacrimal gland pathology in the nonobese diabetic mouse model of Sjögren syndrome. Immunol Cell Biol 2017; 95:684-694. [PMID: 28465508 PMCID: PMC5595634 DOI: 10.1038/icb.2017.38] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 01/01/2023]
Abstract
Sjögren syndrome is an autoimmune disease characterized by targeted destruction of the lacrimal and salivary glands resulting in symptoms of severe ocular and oral dryness. Despite its prevalence, the mechanisms driving autoimmune manifestations are unclear. In patients and in the nonobese diabetic (NOD) mouse model of Sjögren syndrome, lymphocytic infiltrates consist of CD4 and CD8 T cells, although the role of CD8 T cells in disease pathogenesis has been largely unexplored. Here, we evaluated the contribution of CD8 T cells to lacrimal and salivary gland autoimmunity. Within the lacrimal and salivary glands of NOD mice, CD8 T cells were proliferating, expressed an activated phenotype, and produced inflammatory cytokines. Transfer of purified CD8 T cells isolated from the cervical lymph nodes (LNs) of NOD mice into NOD-severe combined immunodeficiency recipients resulted in inflammation of the lacrimal glands, but was not sufficient to cause inflammation of the salivary glands. Lacrimal gland-infiltrating CD8 T cells displayed a cytotoxic phenotype, and epithelial cell damage in the lacrimal glands was observed in recipients of CD8 T cells regardless of the presence of CD4 T cells. Collectively, our results demonstrate that CD8 T cells have a pathogenic role in lacrimal gland autoimmunity. The gland-specific pathogenicity of CD8 T cells makes them a valuable resource to further understand the mechanisms that discriminate lacrimal versus salivary gland autoimmunity and for the development of new therapeutics that target the early stages of disease.
Collapse
Affiliation(s)
- Jennifer Y Barr
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Xiaofang Wang
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - David K Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Scott M Lieberman
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
40
|
Abstract
The immune system provides protection to tissues damaged by infectious microrganisms or physical damage. In autoimmune diseases, the immune system recognizes and attacks its own tissues, i.e., self-destruction. Various agents such as genetic factors and environmental triggers are thought to play a major role in the development of autoimmune diseases. A common feature of all autoimmune diseases is the presence of autoantibodies and inflammation, including mononuclear phagocytes, autoreactive T lymphocytes, and autoantibody producing B cells (plasma cells). It has long been known that B cells produce autoantibodies and, thereby, contribute to the pathogenesis of many autoimmune diseases. Autoimmune diseases can be classified as organ-specific or non-organ specific depending on whether the autoimmune response is directed against a particular tissue or against widespread antigens as in chronic inflammatory autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Both SLE and RA are characterized by the presence of autoantibodies which play a major role in their etiopathogenesis. SLE is characterized by circulating antibodies and immune complex deposition that can trigger an inflammatory damage in organs. RA is a progressive inflammatory disease in which T cells, B cells, and pro-inflammatory cytokines play a key role in its pathophysiology.
Collapse
Affiliation(s)
- Haseeb Ahsan
- a Department of Biochemistry , Jamia Millia Islamia , New Delhi , India
| |
Collapse
|
41
|
Kasela S, Kisand K, Tserel L, Kaleviste E, Remm A, Fischer K, Esko T, Westra HJ, Fairfax BP, Makino S, Knight JC, Franke L, Metspalu A, Peterson P, Milani L. Pathogenic implications for autoimmune mechanisms derived by comparative eQTL analysis of CD4+ versus CD8+ T cells. PLoS Genet 2017; 13:e1006643. [PMID: 28248954 PMCID: PMC5352142 DOI: 10.1371/journal.pgen.1006643] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 03/15/2017] [Accepted: 02/18/2017] [Indexed: 12/28/2022] Open
Abstract
Inappropriate activation or inadequate regulation of CD4+ and CD8+ T cells may contribute to the initiation and progression of multiple autoimmune and inflammatory diseases. Studies on disease-associated genetic polymorphisms have highlighted the importance of biological context for many regulatory variants, which is particularly relevant in understanding the genetic regulation of the immune system and its cellular phenotypes. Here we show cell type-specific regulation of transcript levels of genes associated with several autoimmune diseases in CD4+ and CD8+ T cells including a trans-acting regulatory locus at chr12q13.2 containing the rs1131017 SNP in the RPS26 gene. Most remarkably, we identify a common missense variant in IL27, associated with type 1 diabetes that results in decreased functional activity of the protein and reduced expression levels of downstream IRF1 and STAT1 in CD4+ T cells only. Altogether, our results indicate that eQTL mapping in purified T cells provides novel functional insights into polymorphisms and pathways associated with autoimmune diseases.
Collapse
Affiliation(s)
- Silva Kasela
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Liina Tserel
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Epp Kaleviste
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Anu Remm
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Harm-Jan Westra
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
- Partners Center for Personalized Genetic Medicine, Boston, MA, United States of America
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
| | - Benjamin P. Fairfax
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Seiko Makino
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Julian C. Knight
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| |
Collapse
|
42
|
Baddack-Werncke U, Busch-Dienstfertig M, González-Rodríguez S, Maddila SC, Grobe J, Lipp M, Stein C, Müller G. Cytotoxic T cells modulate inflammation and endogenous opioid analgesia in chronic arthritis. J Neuroinflammation 2017; 14:30. [PMID: 28166793 PMCID: PMC5294766 DOI: 10.1186/s12974-017-0804-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/26/2017] [Indexed: 11/10/2022] Open
Abstract
Background This study examined the development of chronic pain, a cardinal symptom of rheumatoid arthritis (RA), in mice with antigen- and collagen-induced arthritis (ACIA). Since the role of CD8+ T cells in arthritis is controversial, we investigated the consequences of CD8-depletion on arthritis development and opioid modulation of pain in this novel model of chronic autoimmune arthritis. Methods Disease severity in control and CD8-depleted animals was determined by histological assessment of knee-joint sections and measurement of autoantibody formation. Pain was evaluated by measuring mechanical allodynia and thermal hyperalgesia in von Frey and Hargreaves tests, respectively. The production and release of endogenous opioids and inflammatory cytokines was assessed in immunoassays. Results In ACIA, mice display persistent mechanical allodynia and thermal hyperalgesia for more than 2 months after induction of arthritis. The blockade of peripheral opioid receptors with naloxone-methiodide (NLXM) transiently increased thermal hyperalgesia, indicating that endogenous opioid peptides were released in the arthritic joint to inhibit pain. CD8+ T cell depletion did not affect autoantibody formation or severity of joint inflammation, but serum levels of the pro-inflammatory cytokines TNFα and IL-17 were increased. The release of opioid peptides from explanted arthritic knee cells and the NLXM effect were significantly reduced in the absence of CD8+ T cells. Conclusions We have successfully modeled the development of chronic pain, a hallmark of RA, in ACIA. Furthermore, we detected a yet unknown protective role of CD8+ T cells in chronic ACIA since pro-inflammatory cytokines rose and opioid peptide release decreased in the absence of these cells.
Collapse
Affiliation(s)
- Uta Baddack-Werncke
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center of Molecular Medicine (MDC), Robert-Rössle-Strasse 10, 13125, Berlin, Germany.,Current address: DLR project management agency, Department for Health Research, Heinrich-Konen-Str. 1, 53227, Bonn, Germany
| | - Melanie Busch-Dienstfertig
- Department of Anesthesiology and Critical Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Sara González-Rodríguez
- Department of Anesthesiology and Critical Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany. .,Current address: Instituto de Biología Molecular y Celular (IBMC), Av. de la Universidad s/n. Edif. Torregaitán, Elche, 03202, Alicante, Spain.
| | - Santhosh Chandar Maddila
- Department of Anesthesiology and Critical Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.,Current address: Santhosh Nursing Home, Darsi, Prakasam District, Andhra Pradesh, 523247, India
| | - Jenny Grobe
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center of Molecular Medicine (MDC), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Martin Lipp
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center of Molecular Medicine (MDC), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Christoph Stein
- Department of Anesthesiology and Critical Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Gerd Müller
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center of Molecular Medicine (MDC), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| |
Collapse
|
43
|
Joglekar MV, Trivedi PM, Kay TW, Hawthorne WJ, O'Connell PJ, Jenkins AJ, Hardikar AA, Thomas HE. Human islet cells are killed by BID-independent mechanisms in response to FAS ligand. Apoptosis 2016; 21:379-89. [PMID: 26758067 DOI: 10.1007/s10495-016-1212-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cell death via FAS/CD95 can occur either by activation of caspases alone (extrinsic) or by activation of mitochondrial death signalling (intrinsic) depending on the cell type. The BH3-only protein BID is activated in the BCL-2-regulated or mitochondrial apoptosis pathway and acts as a switch between the extrinsic and intrinsic cell death pathways. We have previously demonstrated that islets from BID-deficient mice are protected from FAS ligand-mediated apoptosis in vitro. However, it is not yet known if BID plays a similar role in human beta cell death. We therefore aimed to test the role of BID in human islet cell apoptosis immediately after isolation from human cadaver donors, as well as after de-differentiation in vitro. Freshly isolated human islets or 10-12 day cultured human islet cells exhibited BID transcript knockdown after BID siRNA transfection, however they were not protected from FAS ligand-mediated cell death in vitro as determined by DNA fragmentation analysis using flow cytometry. On the other hand, the same cells transfected with siRNA for FAS-associated via death domain (FADD), a molecule in the extrinsic cell death pathway upstream of BID, showed significant reduction in cell death. De-differentiated islets (human islet-derived progenitor cells) also demonstrated similar results with no difference in cell death after BID knockdown as compared to scramble siRNA transfections. Our results indicate that BID-independent pathways are responsible for FAS-dependent human islet cell death. These results are different from those observed in mouse islets and therefore demonstrate potentially alternate pathways of FAS ligand-induced cell death in human and mouse islet cells.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Prerak M Trivedi
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Thomas W Kay
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Wayne J Hawthorne
- The Centre for Transplant and Renal Research, Westmead Millennium Research Institute, University of Sydney, Westmead, Australia
| | - Philip J O'Connell
- The Centre for Transplant and Renal Research, Westmead Millennium Research Institute, University of Sydney, Westmead, Australia
| | - Alicia J Jenkins
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, NHMRC-Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Melbourne, VIC, 3065, Australia. .,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
44
|
Chabot S, Alvarez F, Amrani A, Djilali-Saiah I. Novel model of double transgenic mouse results in autoimmune diabetes in males. Autoimmunity 2016; 49:397-404. [PMID: 27683954 DOI: 10.1080/08916934.2016.1203907] [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: 10/21/2022]
Abstract
Identifying the type of diabetogenic CD8+ T cells that initiate autoimmune diabetes (AID) is a critical step in designing appropriate strategies for the early detection of beta cell-directed autoimmunity and its progression to diabetes. We generated a novel double transgenic (Tg) mouse model on the naturally diabetes resistant C57Bl/6 background, co-expressing two transgenes including a specific TCR anti-lymphocytic choriomeningitis virus nucleoprotein (LCMV-NP) carried by CD8+ T cells and LCMV-NP (as neo-self antigen) expressed by pancreatic beta cells. The resulting double Tg mouse showed partial thymic deletion of the NP-specific CD8+ T cells. The escaping autoreactive NP-specific CD8+ T cells joining the periphery were activated and gained effector functions. Both male and female mice mounted anti-NP antibodies, but only one-fourth adult males spontaneously developed AID. Significant upregulation of the CD44 and CD122 markers as compared to healthy male and female mice characterized the phenotype of diabetogenic CD8+ T cells in diabetic male mice. We also show that only 10% of these CD8+ T cells expressed programmed death 1 receptor (PD-1). Together, these results suggest that in our double Tg mouse model, Ag-specific effector CD44+CD122+PD-1-CD8+ T cell subpopulation is associated with the pathogenesis of AID.
Collapse
Affiliation(s)
- Sylvie Chabot
- a Gastroenterology, Hepatology and Nutrition Division, CHU Sainte-Justine , Montreal , QC , Canada.,b Department of Microbiology , Infectiology and Immunology, University of Montreal , Montreal , QC , Canada
| | - Fernando Alvarez
- a Gastroenterology, Hepatology and Nutrition Division, CHU Sainte-Justine , Montreal , QC , Canada.,b Department of Microbiology , Infectiology and Immunology, University of Montreal , Montreal , QC , Canada.,c Department of Pediatrics , University of Montreal , Montreal , QC , Canada , and
| | - Abdelaziz Amrani
- d Immunology Division, Department of Pediatrics, University of Sherbrooke , Sherbrooke , QC , Canada
| | - Idriss Djilali-Saiah
- a Gastroenterology, Hepatology and Nutrition Division, CHU Sainte-Justine , Montreal , QC , Canada
| |
Collapse
|
45
|
Wabnitz GH, Balta E, Schindler S, Kirchgessner H, Jahraus B, Meuer S, Samstag Y. The pro-oxidative drug WF-10 inhibits serial killing by primary human cytotoxic T-cells. Cell Death Discov 2016; 2:16057. [PMID: 27551545 PMCID: PMC4979520 DOI: 10.1038/cddiscovery.2016.57] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/05/2016] [Indexed: 12/22/2022] Open
Abstract
Cytotoxic T-cells (CTLs) play an important role in many immune-mediated inflammatory diseases. Targeting cytotoxicity of CTLs would allow to interfere with immune-mediated tissue destruction. Here we demonstrate that WF-10, a pro-oxidative compound, inhibits CTL-mediated cytotoxicity. WF-10 did not influence early steps of target-cell killing, but impaired the ability of CTLs to detach from the initial target cell and to move to a second target cell. This reduced serial killing was accompanied by stronger enrichment of the adhesion molecule LFA-1 in the cytolytic immune synapse. LFA-1 clustering requires activation of the actin-bundling protein L-plastin and was accordingly diminished in L-plastin knockdown cells. Interestingly, WF-10 likely acts through regulating L-plastin: (I) It induced L-plastin activation through phosphorylation leading to enhanced LFA-1-mediated cell adhesion, and, importantly, (II) WF-10 lost its influence on target-cell killing in L-plastin knockdown cells. Finally, we demonstrate that WF-10 can improve immunosuppression by conventional drugs. Thus, while cyclosporine A alone had no significant effect on cytotoxicity of CTLs, a combination of cyclosporine A and WF-10 blocked target-cell killing synergistically. Together, our findings suggest that WF-10 – either alone or in combination with conventional immunosuppressive drugs – may be efficient to control progression of diseases, in which CTLs are crucially involved.
Collapse
Affiliation(s)
- G H Wabnitz
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - E Balta
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - S Schindler
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - H Kirchgessner
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - B Jahraus
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - S Meuer
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| | - Y Samstag
- Institute of Immunology, Ruprecht-Karls-University , Im Neuenheimer Feld 305, Heidelberg D-69120, Germany
| |
Collapse
|
46
|
Zhang N, Zhang J, Cao Z, Deng L, Wu Y, Liu H, Qiu Z. Effect of pulse high-volume hemofiltration on Toll-like receptor in patients with severe sepsis. Exp Ther Med 2016; 12:1377-1382. [PMID: 27588059 PMCID: PMC4998014 DOI: 10.3892/etm.2016.3473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 06/23/2016] [Indexed: 12/13/2022] Open
Abstract
The expression level and prognosis of Toll-like receptor 2 (TLR2) mRNA in peripheral blood mononuclear cells of patients with severe sepsis after applying pulse high-volume hemofiltration (PHVHF) were investigated. Sustained PHVHF treatment was carried out on 40 patients on the basis of conventional treatment for up to 72 h. Acute physiology and chronic health evaluation (APACHE) II scores of patients were compared before and after the treatment. CD4+, CD8+ lymphocyte counts and ratios in the peripheral blood were detected using FASort before and 24 and 48 h of PHVHF treatment. Enzyme-linked immunosorbent assay was adopted to detect tumor necrosis factor-α (TNF-α) and interleukin-10 (IL-10) concentrations in plasma at different time points before and after 24, 48 and 72 h of treatment, while semi-quantitative reverse transcription-polymerase chain reaction technology was used to test TLR2 mRNA expression. After PHVHF treatment, APACHE II, Sequential Organ Failure Assessment scores were decreased (P<0.05). After 72 h of PHVHF treat-ment, TNF-α, IL-10, TLR2 mRNA expression levels in the plasma of patients were significantly decreased compared to before treatment (P<0.05), and the IL-10 / TNF-α ratio was much higher than before treatment (p<0.05). In conclusion, PHVHF can restore the pro-inflammatory/anti-inflammatory balance of the body, thereby improving the overall condition of the patients by removing inflammatory mediators and lowering TLR2 expression of mononuclear cell surface in peripheral blood.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Critical Care Medicine, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Jian Zhang
- Department of Critical Care Medicine, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Zhuo Cao
- Department of Respiratory Medicine, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Liancheng Deng
- Department of Critical Care Medicine, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Yifen Wu
- Department of Critical Care Medicine, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Hong Liu
- Department of Critical Care Medicine, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Zeliang Qiu
- Department of Critical Care Medicine, The Affiliated Zhoupu Hospital of Shanghai University of Medicine & Health Sciences, Shanghai 201318, P.R. China
| |
Collapse
|
47
|
López-Cacho JM, Gallardo S, Posada M, Aguerri M, Calzada D, Mayayo T, Lahoz C, Cárdaba B. Characterization of immune cell phenotypes in adults with autism spectrum disorders. J Investig Med 2016; 64:1179-85. [PMID: 27296457 DOI: 10.1136/jim-2016-000070] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2016] [Indexed: 11/03/2022]
Abstract
Autism spectrum disorders (ASDs) are neurodevelopmental disorders characterized by impairments in verbal and non-verbal communication, impaired social interactions and repetitive behaviors. There is evidence of a link between ASD symptoms and immune dysfunction, but few studies have been performed in adult patients to confirm this. In this work, we used flow cytometry to study immunological differences in peripheral blood mononuclear cells from 59 adult patients and 26 healthy control subjects to identify possible immune cell profiles related with this group of disorders. We analyzed six immune cell subpopulations (ie, B-cells, CD4(+) and CD8(+) T-cells, NK, NKT cells, and monocytes) and their corresponding stages of apoptosis and activation. The most noteworthy results showed that, compared to healthy controls, patients had increased percentages of CD8(+) T-cells and B-cells, and a decrease in the percentage of NKT cells. Regarding CD25 expression, we found overall CD25(+) overexpression, primarily in NK and NKT cells. Apoptosis percentage showed an increasing trend only in monocytes of patients. These data support a link between ASD and immune dysfunction, suggesting that specific cellular phenotypes and/or activation status of immune cells may be relevant in adult ASD.
Collapse
Affiliation(s)
| | - Soledad Gallardo
- Department of Immunology, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain
| | - Manuel Posada
- Institute for Rare Disease Research, Instituto de Salud Carlos III, Biomedical Research Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Miriam Aguerri
- Department of Immunology, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain
| | - David Calzada
- Department of Immunology, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain
| | - Teodoro Mayayo
- Department of Sani-Red SL, Parque Científico de Barcelona, Barcelona, Spain
| | - Carlos Lahoz
- Department of Immunology, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain Biomedical Research Network on Respiratory Diseases, (CIBERES), Madrid, Spain
| | - Blanca Cárdaba
- Department of Immunology, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain Biomedical Research Network on Respiratory Diseases, (CIBERES), Madrid, Spain
| |
Collapse
|
48
|
De Cunto G, Lunghi B, Bartalesi B, Cavarra E, Fineschi S, Ulivieri C, Lungarella G, Lucattelli M. Severe Reduction in Number and Function of Peripheral T Cells Does Not Afford Protection toward Emphysema and Bronchial Remodeling Induced in Mice by Cigarette Smoke. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1814-1824. [PMID: 27157991 DOI: 10.1016/j.ajpath.2016.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/24/2016] [Accepted: 03/04/2016] [Indexed: 11/28/2022]
Abstract
The protein Lck (p56(Lck)) is a Src family tyrosine kinase expressed at all stages of thymocyte development and is required for maturation of T cells. The targeted disruption of Lck gene in mice results in severe block in thymocyte maturation with substantial reduction in the development of CD4(+)CD8(+) thymocytes, severe reduction of peripheral T cells, and disruption of T-cell receptor signaling with defective function of T-cell responses. To investigate the role of T lymphocyte in the development of cigarette smoke-induced pulmonary changes, Lck(-/-) mice and corresponding congenic wild-type mice were chronically exposed to cigarette smoke, and their lungs were analyzed by biochemical, immunologic, and morphometric methods. Smoking mice from both genotypes showed disseminated foci of emphysema and large areas of goblet cell metaplasia in bronchial and bronchiolar epithelium. Morphometric evaluation of lung changes and lung elastin determination confirmed that mice from both genotypes showed the same degree of emphysematous lesions. Thus, cigarette smoke exposure in the presence of severe reduction in number and function of peripheral T cells does not influence the development of pulmonary changes induced by cigarette smoke. The data obtained suggest that innate immunity is a leading actor in the early development of pulmonary changes in smoking mice and that the adaptive immune response may play a role at later stages.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Benedetta Lunghi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Silvia Fineschi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| |
Collapse
|
49
|
Sidney J, Schloss J, Moore C, Lindvall M, Wriston A, Hunt DF, Shabanowitz J, DiLorenzo TP, Sette A. Characterization of the peptide binding specificity of the HLA class I alleles B*38:01 and B*39:06. Immunogenetics 2016; 68:231-6. [PMID: 26754738 PMCID: PMC4760861 DOI: 10.1007/s00251-015-0898-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/30/2015] [Indexed: 01/27/2023]
Abstract
B*38:01 and B*39:06 are present with phenotypic frequencies <2% in the general population, but are of interest as B*39:06 is the B allele most associated with type 1 diabetes susceptibility and 38:01 is most protective. A previous study derived putative main anchor motifs for both alleles based on peptide elution data. The present study has utilized panels of single amino acid substitution peptide libraries to derive detailed quantitative motifs accounting for both primary and secondary influences on peptide binding. From these analyses, both alleles were confirmed to utilize the canonical position 2/C-terminus main anchor spacing. B*38:01 preferentially bound peptides with the positively charged or polar residues H, R, and Q in position 2 and the large hydrophobic residues I, F, L, W, and M at the C-terminus. B*39:06 had a similar preference for R in position 2, but also well-tolerated M, Q, and K. A more dramatic contrast between the two alleles was noted at the C-terminus, where the specificity of B*39:06 was clearly for small residues, with A as most preferred, followed by G, V, S, T, and I. Detailed position-by-position and residue-by-residue coefficient values were generated from the panels to provide detailed quantitative B*38:01 and B*39:06 motifs. It is hoped that these detailed motifs will facilitate the identification of T cell epitopes recognized in the context of two class I alleles associated with dramatically different dispositions towards type 1 diabetes, offering potential avenues for the investigation of the role of CD8 T cells in this disease.
Collapse
Affiliation(s)
- John Sidney
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Jennifer Schloss
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Carrie Moore
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Mikaela Lindvall
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Amanda Wriston
- Department of Chemistry, University of Virginia, Charlottesville, VA, 222904, USA
| | - Donald F Hunt
- Departments of Chemistry and Pathology, University of Virginia, Charlottesville, VA, 222904, USA
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, VA, 222904, USA
| | - Teresa P DiLorenzo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
- Department of Medicine (Division of Endocrinology), Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA.
| |
Collapse
|
50
|
Enhanced Direct Major Histocompatibility Complex Class I Self-Antigen Presentation Induced by Chlamydia Infection. Infect Immun 2015; 84:480-90. [PMID: 26597986 DOI: 10.1128/iai.01254-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/17/2015] [Indexed: 11/20/2022] Open
Abstract
The direct major histocompatibility complex (MHC) class I antigen presentation pathway ensures intracellular peptides are displayed at the cellular surface for recognition of infected or transformed cells by CD8(+) cytotoxic T lymphocytes. Chlamydia spp. are obligate intracellular bacteria and, as such, should be targeted by CD8(+) T cells. It is likely that Chlamydia spp. have evolved mechanisms to avoid the CD8(+) killer T cell responses by interfering with MHC class I antigen presentation. Using a model system of self-peptide presentation which allows for posttranslational control of the model protein's stability, we tested the ability of various Chlamydia species to alter direct MHC class I antigen presentation. Infection of the JY lymphoblastoid cell line limited the accumulation of a model host protein and increased presentation of the model-protein-derived peptides. Enhanced self-peptide presentation was detected only when presentation was restricted to defective ribosomal products, or DRiPs, and total MHC class I levels remained unaltered. Skewed antigen presentation was dependent on a bacterial synthesized component, as evidenced by reversal of the observed phenotype upon preventing bacterial transcription, translation, and the inhibition of bacterial lipooligosaccharide synthesis. These data suggest that Chlamydia spp. have evolved to alter the host antigen presentation machinery to favor presentation of defective and rapidly degraded forms of self-antigen, possibly as a mechanism to diminish the presentation of peptides derived from bacterial proteins.
Collapse
|