1
|
He Z, Zhou Q, Du J, Huang Y, Wu B, Xu Z, Wang C, Cheng X. Integrated single-cell and bulk RNA sequencing reveals CREM is involved in the pathogenesis of ulcerative colitis. Heliyon 2024; 10:e27805. [PMID: 38496850 PMCID: PMC10944264 DOI: 10.1016/j.heliyon.2024.e27805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Background Ulcerative colitis (UC) is an inflammatory bowel disease characterized by persistent colonic inflammation. Here, we performed a systematic analysis to gain better insights into UC pathogenesis. Methods We analyzed two UC-related datasets extracted from the gene expression omnibus database using several bioinformatics tools. The primary cell types and key subgroups of primary cells associated with UC and differentially expressed genes (DEGs) between UC and control samples were identified. The molecular regulation of the key genes was also predicted. The gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses of marker genes of key cell subgroups and model genes were performed. The expression of key enriched genes was validated in 10 clinical samples using real-time quantitative polymerase chain reaction (RT-qPCR). Results Monocytes were identified as the major cell type. Ten differentially expressed marker genes were obtained by intersecting the 3121 DEGs, 38 marker genes in major cell types, and 104 marker genes in key cell subgroups. Four essential genes, associated with immune response, were obtained using support vector machine recursive feature elimination and least absolute shrinkage and selection operator analyses. The four essential genes were highly expressed in Cluster 0 during differentiation. Validation of the four key genes in colonic mucosal biopsy specimens from 10 normal and 10 UC patients revealed that CREM was highly expressed in both the lesion-free sites and lesion sites colonic mucosa of UC patients compared with normal adults. Conclusions We identified CREM involved in UC pathogenesis, which is expected to provide a new therapeutic target for UC.
Collapse
Affiliation(s)
- Zongqi He
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, PR China
| | - Qing Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210004, PR China
| | - Jun Du
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, PR China
| | - Yuyu Huang
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, PR China
| | - Bensheng Wu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, PR China
| | - Zhizhong Xu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, PR China
| | - Chao Wang
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, PR China
| | - Xudong Cheng
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215009, PR China
| |
Collapse
|
2
|
Xiang J, Cao J, Shen J, Wang X, Liang J, Li X, Zhang L, Tang B. Bioinformatics analysis reveals the potential common genes and immune characteristics between atrial fibrillation and periodontitis. J Periodontal Res 2024; 59:104-118. [PMID: 37971162 DOI: 10.1111/jre.13192] [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: 05/07/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND OBJECTIVE Atrial fibrillation (AF) and periodontitis, both classified under chronic inflammatory diseases, share common etiologies, including genetic factors and immune pathways. However, the exact mechanisms are still poorly understood. This study aimed to explore the potential common genes and immune characteristics between AF and periodontitis. METHODS Gene expression datasets for AF and periodontitis were downloaded from the Gene Expression Omnibus (GEO) database. Differential expression analysis was used to identify common genes in the training set. Functional analyses, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, were conducted to elucidate the underlying mechanisms. Hub genes were further screened based on expression levels, receiver operating characteristic (ROC) curves, and least absolute shrinkage and selection operator (LASSO) regression. Then, based on the expression levels and ROC values of the hub genes in the validation set, the target genes were identified. Finally, immune cell infiltration analysis was performed on the AF and periodontitis datasets in the training set using the "CIBERSORT" R package. The relationships between target genes, infiltrating immune cells, and inflammatory factors were also investigated. In addition, AF susceptibility, atrial fibrosis, inflammatory infiltration, and RGS1 protein expression in rat models of periodontitis were assessed through in vivo electrophysiology experiments, Masson's trichrome staining, hematoxylin-eosin staining, immunohistochemistry, and western blotting, respectively. RESULTS A total of 21 common genes were identified between AF and periodontitis among the differentially expressed genes. After evaluating gene expression levels, ROC curves, and LASSO analysis, four significant genes between AF and periodontitis were identified, namely regulator of G-protein signaling 1 (RGS1), annexin A6 (ANXA6), solute carrier family 27 member 6 (SLC27A6), and ficolin 1 (FCN1). Further validation confirmed that RGS1 was the optimal shared target gene for AF and periodontitis. Immune cell infiltration analysis revealed that neutrophils and T cells play an important role in the pathogenesis of both diseases. RGS1 showed a significant positive correlation with activated memory CD4 T cells and gamma-delta T cells and a negative correlation with CD8 T cells and regulatory T cells in both training sets. Moreover, RGS1 was positively correlated with classical pro-inflammatory cytokines IL1β and IL6. In periodontitis rat models, AF susceptibility, atrial fibrosis, and inflammatory infiltration were significantly increased, and RGS1 expression in the atrial tissue was upregulated. CONCLUSION A common gene between AF and periodontitis, RGS1 appears central in linking the two conditions. Immune and inflammatory responses may underlie the interaction between AF and periodontitis.
Collapse
Affiliation(s)
- Jie Xiang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
| | - Jiaru Cao
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
| | - Jun Shen
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
| | - Xiaoyan Wang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
| | - Junqing Liang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
| | - Xinshang Li
- Department of General and Emergency Dentistry, The First Affiliated Hospital (The Affiliated S Tomato Logical Hospital) of Xinjiang Medical University, Xinjiang, Urumqi, China
| | - Ling Zhang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
| | - Baopeng Tang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Urumqi, China
| |
Collapse
|
3
|
Flynn AL, Gans J, Escobedo J, Zhu C, Florescu AM, Shankara S, Madden SL, Kim PS, Pao LI. RGS1 Modulates Autophagic and Metabolic Programs and Is a Critical Mediator of Human Regulatory T Cell Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1656-1668. [PMID: 37850953 DOI: 10.4049/jimmunol.2200402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/27/2023] [Indexed: 10/19/2023]
Abstract
Regulatory T cells (Tregs) are critical mediators of immune tolerance and play a diametric role in cancer and autoimmunity. Tumor-infiltrating Tregs are often associated with poor prognosis in solid tumors because their enrichment in the tumor microenvironment contributes to immunosuppression. Conversely, dysregulation in the Treg compartment can disrupt self-tolerance, leading to autoimmunity. In the present study, we describe what is, to our knowledge, a novel regulator of Tregs, the GTPase activator regulator of G protein 1 (RGS1), demonstrating that RGS1-deficient human Tregs show downregulation of Treg-associated genes and are less immunosuppressive. These RGS1-deficient Tregs exhibit perturbations to the FOXP3-c-MYC transcriptional axis and downstream metabolic and autophagy programs by shifting their energy demands toward glycolysis and rendering them less autophagic. Taken together, RGS1 may serve as an apical node of Treg function by regulating the FOXP3-c-MYC transcriptional axis, thereby providing a therapeutic rationale for targeting RGS1 for treatment of cancer and autoimmune diseases.
Collapse
Affiliation(s)
| | - Joseph Gans
- Department of Translational Sciences, Sanofi, Cambridge, MA
| | | | - Cheng Zhu
- Department of Translational Sciences, Sanofi, Cambridge, MA
| | | | | | | | - Peter S Kim
- Department of Oncology, Sanofi, Cambridge, MA
| | - Lily I Pao
- Department of Oncology, Sanofi, Cambridge, MA
| |
Collapse
|
4
|
Chasman DA, Welch Schwartz R, Vazquez J, Chavarria M, Jenkins ET, Lopez GE, Tyler CT, Stanic AK, Ong IM. Proteogenomic and V(D)J Analysis of Human Decidual T Cells Highlights Unique Transcriptional Programming and Clonal Distribution. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:154-162. [PMID: 37195197 PMCID: PMC10330249 DOI: 10.4049/jimmunol.2200061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/25/2023] [Indexed: 05/18/2023]
Abstract
Immunological tolerance toward the semiallogeneic fetus is one of many maternal adaptations required for a successful pregnancy. T cells are major players of the adaptive immune system and balance tolerance and protection at the maternal-fetal interface; however, their repertoire and subset programming are still poorly understood. Using emerging single-cell RNA sequencing technologies, we simultaneously obtained transcript, limited protein, and receptor repertoire at the single-cell level, from decidual and matched maternal peripheral human T cells. The decidua maintains a tissue-specific distribution of T cell subsets compared with the periphery. We find that decidual T cells maintain a unique transcriptome programming, characterized by restraint of inflammatory pathways by overexpression of negative regulators (DUSP, TNFAIP3, ZFP36) and expression of PD-1, CTLA-4, TIGIT, and LAG3 in some CD8 clusters. Finally, analyzing TCR clonotypes demonstrated decreased diversity in specific decidual T cell populations. Overall, our data demonstrate the power of multiomics analysis in revealing regulation of fetal-maternal immune coexistence.
Collapse
Affiliation(s)
- Deborah A. Chasman
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
- Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI
| | - Rene Welch Schwartz
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
- Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Jessica Vazquez
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Melina Chavarria
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Eryne T. Jenkins
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Gladys E. Lopez
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Chanel T. Tyler
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Aleksandar K. Stanic
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
| | - Irene M. Ong
- Departments of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI
- Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
5
|
Chen J, Liu K, Luo Y, Kang M, Wang J, Chen G, Qi J, Wu W, Wang B, Han Y, Shi L, Wang K, Han X, Ma X, Liu W, Ding Y, Wang L, Liang H, Wang L, Chen J. Single-Cell Profiling of Tumor Immune Microenvironment Reveals Immune Irresponsiveness in Gastric Signet-Ring Cell Carcinoma. Gastroenterology 2023; 165:88-103. [PMID: 36921674 DOI: 10.1053/j.gastro.2023.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND & AIMS Gastric cancer (GC) is a major cancer type characterized by high heterogeneity in both tumor cells and the tumor immune microenvironment (TIME). One intractable GC subtype is gastric signet-ring cell carcinoma (GSRCC), which is associated with poor prognosis. However, it remains unclear what the GSRCC TIME characteristics are and how these characteristics may contribute to clinical outcomes. METHODS We enrolled 32 patients with advanced GC of diverse subtypes and profiled their TIME using an immune-targeted single-cell profiling strategy, including (1) immune-targeted single-cell RNA sequencing (n = 20 patients) and (2) protein expression profiling by a targeted antibody panel for mass cytometry (n = 12 patients). We also generated matched V(D)J (variable, diversity, and joining gene segments) sequencing of T and B cells along CD45+ immunocytes. RESULTS We found that compared to non-GSRCC, the GSRCC TIME appears to be quiescent, where both CD4+ and CD8+ T cells are difficult to be mobilized, which further impairs the proper functions of B cells. CXCL13, mainly produced by follicular helper T cells, T helper type 17, and exhausted CD8+ T cells, is a central coordinator of this transformation. We show that CXCL13 expression can predict the response to immune checkpoint blockade in GC patients, which may be related to its effects on tertiary lymphoid structures. CONCLUSIONS Our study provides a comprehensive molecular portrait of immune cell compositions and cell states in advanced GC patients, highlighting adaptive immune irresponsiveness in GSRCC and a mediator role of CXCL13 in TIME. Our targeted single-cell transcriptomic and proteomic profiling represents a powerful approach for TIME-oriented translational research.
Collapse
Affiliation(s)
- Jing Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kuai Liu
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China
| | - Yikai Luo
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, Texas; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Muxing Kang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guofeng Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Qi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenxuan Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Beidi Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaxuan Han
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Le Shi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefan Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaying Han
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojing Ma
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuan Ding
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liangjing Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, Texas.
| | - Lie Wang
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Jian Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Adzibolosu N, Alvero AB, Ali-Fehmi R, Gogoi R, Corey L, Tedja R, Chehade H, Gogoi V, Morris R, Anderson M, Vitko J, Lam C, Craig DB, Draghici S, Rutherford T, Mor G. Immunological modifications following chemotherapy are associated with delayed recurrence of ovarian cancer. Front Immunol 2023; 14:1204148. [PMID: 37435088 PMCID: PMC10331425 DOI: 10.3389/fimmu.2023.1204148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction Ovarian cancer recurs in most High Grade Serous Ovarian Cancer (HGSOC) patients, including initial responders, after standard of care. To improve patient survival, we need to identify and understand the factors contributing to early or late recurrence and therapeutically target these mechanisms. We hypothesized that in HGSOC, the response to chemotherapy is associated with a specific gene expression signature determined by the tumor microenvironment. In this study, we sought to determine the differences in gene expression and the tumor immune microenvironment between patients who show early recurrence (within 6 months) compared to those who show late recurrence following chemotherapy. Methods Paired tumor samples were obtained before and after Carboplatin and Taxol chemotherapy from 24 patients with HGSOC. Bioinformatic transcriptomic analysis was performed on the tumor samples to determine the gene expression signature associated with differences in recurrence pattern. Gene Ontology and Pathway analysis was performed using AdvaitaBio's iPathwayGuide software. Tumor immune cell fractions were imputed using CIBERSORTx. Results were compared between late recurrence and early recurrence patients, and between paired pre-chemotherapy and post-chemotherapy samples. Results There was no statistically significant difference between early recurrence or late recurrence ovarian tumors pre-chemotherapy. However, chemotherapy induced significant immunological changes in tumors from late recurrence patients but had no impact on tumors from early recurrence patients. The key immunological change induced by chemotherapy in late recurrence patients was the reversal of pro-tumor immune signature. Discussion We report for the first time, the association between immunological modifications in response to chemotherapy and the time of recurrence. Our findings provide novel opportunities to ultimately improve ovarian cancer patient survival.
Collapse
Affiliation(s)
- Nicholas Adzibolosu
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Ayesha B. Alvero
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Rouba Ali-Fehmi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Radhika Gogoi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Logan Corey
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Roslyn Tedja
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Hussein Chehade
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Center of Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Vir Gogoi
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Robert Morris
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Matthew Anderson
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Julie Vitko
- Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Clarissa Lam
- Department of Gynecologic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Douglas B. Craig
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, United States
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Sorin Draghici
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, United States
- Advaita Corporation, Ann Arbor, MI, United States
- Division of Information and Intelligent Systems, Directorate for Computer and Information Science and Engineering, National Science Foundation, Alexandria, VA, United States
| | - Thomas Rutherford
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Gil Mor
- C. S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
7
|
Wu Y, Gettler K, Kars ME, Giri M, Li D, Bayrak CS, Zhang P, Jain A, Maffucci P, Sabic K, Van Vleck T, Nadkarni G, Denson LA, Ostrer H, Levine AP, Schiff ER, Segal AW, Kugathasan S, Stenson PD, Cooper DN, Philip Schumm L, Snapper S, Daly MJ, Haritunians T, Duerr RH, Silverberg MS, Rioux JD, Brant SR, McGovern DPB, Cho JH, Itan Y. Identifying high-impact variants and genes in exomes of Ashkenazi Jewish inflammatory bowel disease patients. Nat Commun 2023; 14:2256. [PMID: 37080976 PMCID: PMC10119186 DOI: 10.1038/s41467-023-37849-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 04/03/2023] [Indexed: 04/22/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a group of chronic digestive tract inflammatory conditions whose genetic etiology is still poorly understood. The incidence of IBD is particularly high among Ashkenazi Jews. Here, we identify 8 novel and plausible IBD-causing genes from the exomes of 4453 genetically identified Ashkenazi Jewish IBD cases (1734) and controls (2719). Various biological pathway analyses are performed, along with bulk and single-cell RNA sequencing, to demonstrate the likely physiological relatedness of the novel genes to IBD. Importantly, we demonstrate that the rare and high impact genetic architecture of Ashkenazi Jewish adult IBD displays significant overlap with very early onset-IBD genetics. Moreover, by performing biobank phenome-wide analyses, we find that IBD genes have pleiotropic effects that involve other immune responses. Finally, we show that polygenic risk score analyses based on genome-wide high impact variants have high power to predict IBD susceptibility.
Collapse
Affiliation(s)
- Yiming Wu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kyle Gettler
- Department of Genetics, Yale University, New Haven, CT, USA
| | - Meltem Ece Kars
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mamta Giri
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dalin Li
- Translational Genomics Unit, F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cigdem Sevim Bayrak
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY, USA
| | - Aayushee Jain
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick Maffucci
- Immunology Institute, Graduate School, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Ksenija Sabic
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tielman Van Vleck
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lee A Denson
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Harry Ostrer
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, USA
| | - Adam P Levine
- Division of Medicine, University College London (UCL), London, UK
- Research Department of Pathology, University College London (UCL), London, UK
| | - Elena R Schiff
- Division of Medicine, University College London (UCL), London, UK
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Anthony W Segal
- Division of Medicine, University College London (UCL), London, UK
| | | | - Peter D Stenson
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - David N Cooper
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - L Philip Schumm
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Scott Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Oncology Boston Children's Hospital, Boston, MA, USA
| | - Mark J Daly
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Talin Haritunians
- Translational Genomics Unit, F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Richard H Duerr
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Mark S Silverberg
- Inflammatory Bowel Disease Centre, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - John D Rioux
- Research Center, Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Steven R Brant
- Division of Gastroenterology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
- Meyerhoff Inflammatory Bowel Disease Center, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dermot P B McGovern
- Translational Genomics Unit, F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Judy H Cho
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
8
|
Krieg PF, Sonner JK, Kurelic R, Engler JB, Scharenberg MF, Bauer S, Nikolaev VO, Friese MA. GPR52 regulates cAMP in T cells but is dispensable for encephalitogenic responses. Front Immunol 2023; 13:1113348. [PMID: 36761164 PMCID: PMC9902724 DOI: 10.3389/fimmu.2022.1113348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/31/2022] [Indexed: 01/25/2023] Open
Abstract
G-protein coupled receptors (GPCR) regulate 3',5'-cyclic adenosine monophosphate (cAMP) levels in T cells. cAMP as ubiquitous second messenger is crucial for adequate physiology of T cells by mediating effector T cell (Teff) function as well as regulatory T cell (Treg)-mediated immunosuppression. Several GPCRs have been identified to be crucial for Teff and Treg function. However, the role of the orphan, constitutively active Gs-coupled GPCR GPR52 is unknown. Here we show that GPR52 regulates cAMP levels in T cells but does not affect T cell function. We found that stimulation of transfected HEK cells or primary T cells with a GPR52 agonist results in a rise of intracellular cAMP. However, neither Gpr52 deficiency nor pharmacological modulation of GPR52 by antagonists or agonists affected T cell activation, differentiation, and proliferation or Treg-mediated immunosuppression. Moreover, Gpr52 deletion did not modify the clinical disease course of experimental autoimmune encephalomyelitis (EAE). Our results demonstrate that a modulation of cAMP levels in T cells does not inevitably result in altered T cell function. While we could not identify an obvious role of GPR52 in in vitro T cell assays and in vivo CNS autoimmunity, it might regulate T cell function in a different context or affect the function of other GPR52-expressing cells.
Collapse
Affiliation(s)
- Paula F. Krieg
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K. Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roberta Kurelic
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marlena F. Scharenberg
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Manuel A. Friese,
| |
Collapse
|
9
|
von Werdt D, Gungor B, Barreto de Albuquerque J, Gruber T, Zysset D, Kwong Chung CKC, Corrêa-Ferreira A, Berchtold R, Page N, Schenk M, Kehrl JH, Merkler D, Imhof BA, Stein JV, Abe J, Turchinovich G, Finke D, Hayday AC, Corazza N, Mueller C. Regulator of G-protein signaling 1 critically supports CD8 + T RM cell-mediated intestinal immunity. Front Immunol 2023; 14:1085895. [PMID: 37153600 PMCID: PMC10158727 DOI: 10.3389/fimmu.2023.1085895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/13/2023] [Indexed: 05/09/2023] Open
Abstract
Members of the Regulator of G-protein signaling (Rgs) family regulate the extent and timing of G protein signaling by increasing the GTPase activity of Gα protein subunits. The Rgs family member Rgs1 is one of the most up-regulated genes in tissue-resident memory (TRM) T cells when compared to their circulating T cell counterparts. Functionally, Rgs1 preferentially deactivates Gαq, and Gαi protein subunits and can therefore also attenuate chemokine receptor-mediated immune cell trafficking. The impact of Rgs1 expression on tissue-resident T cell generation, their maintenance, and the immunosurveillance of barrier tissues, however, is only incompletely understood. Here we report that Rgs1 expression is readily induced in naïve OT-I T cells in vivo following intestinal infection with Listeria monocytogenes-OVA. In bone marrow chimeras, Rgs1 -/- and Rgs1 +/+ T cells were generally present in comparable frequencies in distinct T cell subsets of the intestinal mucosa, mesenteric lymph nodes, and spleen. After intestinal infection with Listeria monocytogenes-OVA, however, OT-I Rgs1 +/+ T cells outnumbered the co-transferred OT-I Rgs1- /- T cells in the small intestinal mucosa already early after infection. The underrepresentation of the OT-I Rgs1 -/- T cells persisted to become even more pronounced during the memory phase (d30 post-infection). Remarkably, upon intestinal reinfection, mice with intestinal OT-I Rgs1 +/+ TRM cells were able to prevent the systemic dissemination of the pathogen more efficiently than those with OT-I Rgs1 -/- TRM cells. While the underlying mechanisms are not fully elucidated yet, these data thus identify Rgs1 as a critical regulator for the generation and maintenance of tissue-resident CD8+ T cells as a prerequisite for efficient local immunosurveillance in barrier tissues in case of reinfections with potential pathogens.
Collapse
Affiliation(s)
- Diego von Werdt
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Bilgi Gungor
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Thomas Gruber
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Daniel Zysset
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Cheong K. C. Kwong Chung
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Gastrointestinal Health, Immunology, Nestlé Research, Lausanne, Switzerland
| | - Antonia Corrêa-Ferreira
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Regina Berchtold
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Nicolas Page
- Department of Pathology, Division of Clinical Pathology, University & University Hospitals of Geneva, Geneva, Switzerland
| | - Mirjam Schenk
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - John H. Kehrl
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Doron Merkler
- Department of Pathology, Division of Clinical Pathology, University & University Hospitals of Geneva, Geneva, Switzerland
| | - Beat A. Imhof
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Pathology and Immunology, Centre Medical Universitaire, University of Geneva, Geneva, Switzerland
| | - Jens V. Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Gleb Turchinovich
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Finke
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
| | - Adrian C. Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Nadia Corazza
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- *Correspondence: Christoph Mueller, ; Nadia Corazza,
| | - Christoph Mueller
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Biomedicine, and University Children’s Hospital Basel, University of Basel, Basel, Switzerland
- *Correspondence: Christoph Mueller, ; Nadia Corazza,
| |
Collapse
|
10
|
Xue W, He W, Yan M, Zhao H, Pi J. Exploring Shared Biomarkers of Myocardial Infarction and Alzheimer's Disease via Single-Cell/Nucleus Sequencing and Bioinformatics Analysis. J Alzheimers Dis 2023; 96:705-723. [PMID: 37840493 PMCID: PMC10657707 DOI: 10.3233/jad-230559] [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] [Accepted: 09/04/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Patients are at increased risk of dementia, including Alzheimer's disease (AD), after myocardial infarction (MI), but the biological link between MI and AD is unclear. OBJECTIVE To understand the association between the pathogenesis of MI and AD and identify common biomarkers of both diseases. METHODS Using public databases, we identified common biomarkers of MI and AD. Least absolute shrinkage and selection operator (LASSO) regression and protein-protein interaction (PPI) network were performed to further screen hub biomarkers. Functional enrichment analyses were performed on the hub biomarkers. Single-cell/nucleus analysis was utilized to further analyze the hub biomarkers at the cellular level in carotid atherosclerosis and AD datasets. Motif enrichment analysis was used to screen key transcription factors. RESULTS 26 common differentially expressed genes were screened between MI and AD. Function enrichment analyses showed that these differentially expressed genes were mainly associated with inflammatory pathways. A key gene, Regulator of G-protein Signaling 1 (RGS1), was obtained by LASSO regression and PPI network. RGS1 was confirmed to mainly express in macrophages and microglia according to single-cell/nucleus analysis. The difference in expression of RGS1 in macrophages and microglia between disease groups and controls was statistically significant (p < 0.0001). The expression of RGS1 in the disease groups was upregulated with the differentiation of macrophages and microglia. RelA was a key transcription factor regulating RGS1. CONCLUSION Macrophages and microglia are involved in the inflammatory response of MI and AD. RGS1 may be a key biomarker in this process.
Collapse
Affiliation(s)
- Weiqi Xue
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weifeng He
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mengyuan Yan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huanyi Zhao
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianbin Pi
- Department of Cardiovascular Disease, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| |
Collapse
|
11
|
Notarbartolo S, Abrignani S. Human T lymphocytes at tumor sites. Semin Immunopathol 2022; 44:883-901. [PMID: 36385379 PMCID: PMC9668216 DOI: 10.1007/s00281-022-00970-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/14/2022] [Indexed: 12/15/2022]
Abstract
CD4+ and CD8+ T lymphocytes mediate most of the adaptive immune response against tumors. Naïve T lymphocytes specific for tumor antigens are primed in lymph nodes by dendritic cells. Upon activation, antigen-specific T cells proliferate and differentiate into effector cells that migrate out of peripheral blood into tumor sites in an attempt to eliminate cancer cells. After accomplishing their function, most effector T cells die in the tissue, while a small fraction of antigen-specific T cells persist as long-lived memory cells, circulating between peripheral blood and lymphoid tissues, to generate enhanced immune responses when re-encountering the same antigen. A subset of memory T cells, called resident memory T (TRM) cells, stably resides in non-lymphoid peripheral tissues and may provide rapid immunity independently of T cells recruited from blood. Being adapted to the tissue microenvironment, TRM cells are potentially endowed with the best features to protect against the reemergence of cancer cells. However, when tumors give clinical manifestation, it means that tumor cells have evaded immune surveillance, including that of TRM cells. Here, we review the current knowledge as to how TRM cells are generated during an immune response and then maintained in non-lymphoid tissues. We then focus on what is known about the role of CD4+ and CD8+ TRM cells in antitumor immunity and their possible contribution to the efficacy of immunotherapy. Finally, we highlight some open questions in the field and discuss how new technologies may help in addressing them.
Collapse
Affiliation(s)
- Samuele Notarbartolo
- INGM, Istituto Nazionale Genetica Molecolare "Romeo Ed Enrica Invernizzi", Milan, Italy.
| | - Sergio Abrignani
- INGM, Istituto Nazionale Genetica Molecolare "Romeo Ed Enrica Invernizzi", Milan, Italy.
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
12
|
Fardoos R, Nyquist SK, Asowata OE, Kazer SW, Singh A, Ngoepe A, Giandhari J, Mthabela N, Ramjit D, Singh S, Karim F, Buus S, Anderson F, Porterfield JZ, Sibiya AL, Bipath R, Moodley K, Kuhn W, Berger B, Nguyen S, de Oliveira T, Ndung’u T, Goulder P, Shalek AK, Leslie A, Kløverpris HN. HIV specific CD8 + T RM-like cells in tonsils express exhaustive signatures in the absence of natural HIV control. Front Immunol 2022; 13:912038. [PMID: 36330531 PMCID: PMC9623418 DOI: 10.3389/fimmu.2022.912038] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Lymphoid tissues are an important HIV reservoir site that persists in the face of antiretroviral therapy and natural immunity. Targeting these reservoirs by harnessing the antiviral activity of local tissue-resident memory (TRM) CD8+ T-cells is of great interest, but limited data exist on TRM-like cells within lymph nodes of people living with HIV (PLWH). Here, we studied tonsil CD8+ T-cells obtained from PLWH and uninfected controls from South Africa. We show that these cells are preferentially located outside the germinal centers (GCs), the main reservoir site for HIV, and display a low cytolytic and a transcriptionally TRM-like profile distinct from blood CD8+ T-cells. In PLWH, CD8+ TRM-like cells are expanded and adopt a more cytolytic, activated, and exhausted phenotype not reversed by antiretroviral therapy (ART). This phenotype was enhanced in HIV-specific CD8+ T-cells from tonsils compared to matched blood suggesting a higher antigen burden in tonsils. Single-cell transcriptional and clonotype resolution showed that these HIV-specific CD8+ T-cells in the tonsils express heterogeneous signatures of T-cell activation, clonal expansion, and exhaustion ex-vivo. Interestingly, this signature was absent in a natural HIV controller, who expressed lower PD-1 and CXCR5 levels and reduced transcriptional evidence of T-cell activation, exhaustion, and cytolytic activity. These data provide important insights into lymphoid tissue-derived HIV-specific CD8+ TRM-like phenotypes in settings of HIV remission and highlight their potential for immunotherapy and targeting of the HIV reservoirs.
Collapse
Affiliation(s)
- Rabiah Fardoos
- Africa Health Research Institute (AHRI), Durban, South Africa
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sarah K. Nyquist
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | | - Samuel W. Kazer
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Alveera Singh
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Abigail Ngoepe
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Jennifer Giandhari
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Dirhona Ramjit
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Samita Singh
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Farina Karim
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Søren Buus
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Frank Anderson
- Discipline of General Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - J. Zachary Porterfield
- Africa Health Research Institute (AHRI), Durban, South Africa
- Department of Otolaryngology-Head & Neck Surgery, Division of Infectious Diseases, University of Kentucky, Lexington, KY, United States
- Department of Microbiology, Immunology and Molecular Genetics, - Division of Infectious Diseases, University of Kentucky, Lexington, KY, United States
- Department of Internal Medicine - Division of Infectious Diseases, University of Kentucky, Lexington, KY, United States
| | - Andile L. Sibiya
- Department of Otorhinolaryngology & Head & Neck Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Rishan Bipath
- Department of Otorhinolaryngology, King Edward VIII hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Kumeshan Moodley
- Department of Ear Nose and Throat, General Justice Gizenga Mpanza Regional Hospital (Stanger Hospital), University of KwaZulu-Natal, Durban, South Africa
| | - Warren Kuhn
- Department of Otorhinolaryngology & Head & Neck Surgery, Inkosi Albert Luthuli Central Hospital, University of KwaZulu-Natal, Durban, South Africa
- Department of Ear Nose and Throat, General Justice Gizenga Mpanza Regional Hospital (Stanger Hospital), University of KwaZulu-Natal, Durban, South Africa
| | - Bonnie Berger
- Computer Science & Artificial Intelligence Lab and Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Son Nguyen
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Tulio de Oliveira
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Thumbi Ndung’u
- Africa Health Research Institute (AHRI), Durban, South Africa
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu Natal, Durban, South Africa
- University College London, Division of Infection and Immunity, London, United Kingdom
| | - Philip Goulder
- Africa Health Research Institute (AHRI), Durban, South Africa
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu Natal, Durban, South Africa
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Alex K. Shalek
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, United States
- Ragon Institute of MGH, Harvard, Cambridge, MA, United States
| | - Alasdair Leslie
- Africa Health Research Institute (AHRI), Durban, South Africa
- University College London, Division of Infection and Immunity, London, United Kingdom
| | - Henrik N. Kløverpris
- Africa Health Research Institute (AHRI), Durban, South Africa
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- University College London, Division of Infection and Immunity, London, United Kingdom
| |
Collapse
|
13
|
Pawlak M, DeTomaso D, Schnell A, Meyer Zu Horste G, Lee Y, Nyman J, Dionne D, Regan BML, Singh V, Delorey T, Schramm MA, Wang C, Wallrapp A, Burkett PR, Riesenfeld SJ, Anderson AC, Regev A, Xavier RJ, Yosef N, Kuchroo VK. Induction of a colitogenic phenotype in Th1-like cells depends on interleukin-23 receptor signaling. Immunity 2022; 55:1663-1679.e6. [PMID: 36070768 PMCID: PMC9808605 DOI: 10.1016/j.immuni.2022.08.007] [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: 03/09/2022] [Revised: 06/19/2022] [Accepted: 08/11/2022] [Indexed: 01/14/2023]
Abstract
Interleukin-23 receptor plays a critical role in inducing inflammation and autoimmunity. Here, we report that Th1-like cells differentiated in vitro with IL-12 + IL-21 showed similar IL-23R expression to that of pathogenic Th17 cells using eGFP reporter mice. Fate mapping established that these cells did not transition through a Th17 cell state prior to becoming Th1-like cells, and we observed their emergence in vivo in the T cell adoptive transfer colitis model. Using IL-23R-deficient Th1-like cells, we demonstrated that IL-23R was required for the development of a highly colitogenic phenotype. Single-cell RNA sequencing analysis of intestinal T cells identified IL-23R-dependent genes in Th1-like cells that differed from those expressed in Th17 cells. The perturbation of one of these regulators (CD160) in Th1-like cells inhibited the induction of colitis. We thus uncouple IL-23R as a purely Th17 cell-specific factor and implicate IL-23R signaling as a pathogenic driver in Th1-like cells inducing tissue inflammation.
Collapse
Affiliation(s)
- Mathias Pawlak
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David DeTomaso
- Department of Electrical Engineering and Computer Sciences and Center for Computational Biology, UC Berkeley, Berkeley, CA 94720, USA
| | - Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gerd Meyer Zu Horste
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Youjin Lee
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jackson Nyman
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brianna M L Regan
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vasundhara Singh
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Toni Delorey
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Markus A Schramm
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Chao Wang
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Antonia Wallrapp
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Patrick R Burkett
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Samantha J Riesenfeld
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ana C Anderson
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ramnik J Xavier
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, MGH, Boston, MA 02114, USA.
| | - Nir Yosef
- Department of Electrical Engineering and Computer Sciences and Center for Computational Biology, UC Berkeley, Berkeley, CA 94720, USA; Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
14
|
Wei W, Li J, Liu X, Pan W, Wang M, Li J, Yue Y, Hao L. Inhibition of RGS10 Aggravates Periapical Periodontitis via Upregulation of the NF-κB Pathway. J Endod 2022; 48:1308-1318.e5. [PMID: 36041584 DOI: 10.1016/j.joen.2022.07.009] [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: 05/08/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Periapical periodontitis develops due to the interplay between root canal microorganisms and host defenses. The mechanism underlying the pathogenesis of periapical periodontitis remains unclear. Regulator of G protein signaling protein 10 (RGS10) has been suggested to play a role in regulating inflammation. This study explored the potential regulatory effects of RGS10 on periapical periodontitis and the pro-inflammatory pathway of NF-κB. METHODS Disease models of periapical inflammation in mice were established, and adenovirus-associated virus (AAV) was used to inhibit RGS10 expression. Periapical lesions were detected using microcomputed tomography. Quantitative real-time PCR (qRT-PCR), western blotting (WB), enzyme-linked immunosorbent assay (ELISA), enzyme activity staining of tartrate-resistant acid phosphatase, and immunohistochemistry were conducted to assess the role of RGS10 expression on NF-κB pro-inflammatory signaling, OPG, RANKL, and osteoclasts in the periapical regions of each group. TNFα was used to stimulate L929 cells alone or with small interfering RNA (siRNA). To assess the expression of associated molecules, WB, immunofluorescence, qRT-PCR, and ELISA were performed. RESULTS RGS10 inhibition increased alveolar bone destruction in periapical periodontitis lesions and substantially enhanced the NF-κB pro-inflammatory signaling pathway activation level. Furthermore, RGS10 inhibition upregulated the ratio of OPG/RANKL and the maturation of osteoclasts during alveolar bone resorption. L929 cell TNFα stimulation and siRNA transfection confirmed these in vivo results. CONCLUSION RGS10 negatively regulates NF-κB pro-inflammatory signaling in periapical periodontitis and participates in bone remodeling. Therefore, RGS10 is a promising treatment option for long-term chronic periapical inflammation and may be a new target for the artificial regulation of inflammation.
Collapse
Affiliation(s)
- Wei Wei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Jiaxin Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Xinran Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Weiyi Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Min Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Jinle Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Yuan Yue
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China.
| | - Liang Hao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China.
| |
Collapse
|
15
|
Tu E, McGlinchey K, Wang J, Martin P, Ching SL, Floc’h N, Kurasawa J, Starrett JH, Lazdun Y, Wetzel L, Nuttall B, Ng FS, Coffman KT, Smith PD, Politi K, Cooper ZA, Streicher K. Anti-PD-L1 and anti-CD73 combination therapy promotes T cell response to EGFR-mutated NSCLC. JCI Insight 2022; 7:e142843. [PMID: 35132961 PMCID: PMC8855814 DOI: 10.1172/jci.insight.142843] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
Treatment with anti-PD-1 and anti-PD-L1 therapies has shown durable clinical benefit in non-small cell lung cancer (NSCLC). However, patients with NSCLC with epidermal growth factor receptor (EGFR) mutations do not respond as well to treatment as patients without an EGFR mutation. We show that EGFR-mutated NSCLC expressed higher levels of CD73 compared with EGFR WT tumors and that CD73 expression was regulated by EGFR signaling. EGFR-mutated cell lines were significantly more resistant to T cell killing compared with WT cell lines through suppression of T cell proliferation and function. In a xenograft mouse model of EGFR-mutated NSCLC, neither anti-PD-L1 nor anti-CD73 antibody alone inhibited tumor growth compared with the isotype control. In contrast, the combination of both antibodies significantly inhibited tumor growth, increased the number of tumor-infiltrating CD8+ T cells, and enhanced IFN-γ and TNF-α production of these T cells. Consistently, there were increases in gene expression that corresponded to inflammation and T cell function in tumors treated with the combination of anti-PD-L1 and anti-CD73. Together, these results further support the combination of anti-CD73 and anti-PD-L1 therapies in treating EGFR-mutated NSCLC, while suggesting that increased T cell activity may play a role in response to therapy.
Collapse
Affiliation(s)
| | - Kelly McGlinchey
- Research Early Oncology, AstraZeneca, Gaithersburg, Maryland, USA
| | | | | | | | - Nicolas Floc’h
- Oncology R&D, Bioscience, AstraZeneca, Cambridge, United Kingdom
| | - James Kurasawa
- Biologics Engineering, AstraZeneca, Gaithersburg, Maryland, USA
| | | | | | - Leslie Wetzel
- Research Early Oncology, AstraZeneca, Gaithersburg, Maryland, USA
| | | | | | | | - Paul D. Smith
- Oncology R&D, Bioscience, AstraZeneca, Cambridge, United Kingdom
| | - Katerina Politi
- Department of Pathology and Medicine, Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | | | | |
Collapse
|
16
|
Arnone D, Vallier M, Hergalant S, Chabot C, Ndiaye NC, Moulin D, Aignatoaei AM, Alberto JM, Louis H, Boulard O, Mayeur C, Dreumont N, Peuker K, Strigli A, Zeissig S, Hansmannel F, Chamaillard M, Kökten T, Peyrin-Biroulet L. Long-Term Overconsumption of Fat and Sugar Causes a Partially Reversible Pre-inflammatory Bowel Disease State. Front Nutr 2021; 8:758518. [PMID: 34869528 PMCID: PMC8637418 DOI: 10.3389/fnut.2021.758518] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022] Open
Abstract
Nutrition appears to be an important environmental factor involved in the onset of inflammatory bowel diseases (IBD) through yet poorly understood biological mechanisms. Most studies focused on fat content in high caloric diets, while refined sugars represent up to 40% of caloric intake within industrialized countries and contribute to the growing epidemics of inflammatory diseases. Herein we aim to better understand the impact of a high-fat-high-sucrose diet on intestinal homeostasis in healthy conditions and the subsequent colitis risk. We investigated the early events and the potential reversibility of high caloric diet-induced damage in mice before experimental colitis. C57BL/6 mice were fed with a high-fat or high-fat high-sucrose or control diet before experimental colitis. In healthy mice, a high-fat high-sucrose diet induces a pre-IBD state characterized by gut microbiota dysbiosis with a total depletion of bacteria belonging to Barnesiella that is associated with subclinical endoscopic lesions. An overall down-regulation of the colonic transcriptome converged with broadly decreased immune cell populations in the mesenteric lymph nodes leading to the inability to respond to tissue injury. Such in-vivo effects on microbiome and transcriptome were partially restored when returning to normal chow. Long-term consumption of diet enriched in sucrose and fat predisposes mice to colitis. This enhanced risk is preceded by gut microbiota dysbiosis and transcriptional reprogramming of colonic genes related to IBD. Importantly, diet-induced transcriptome and microbiome disturbances are partially reversible after switching back to normal chow with persistent sequelae that may contribute to IBD predisposition in the general population.
Collapse
Affiliation(s)
- Djésia Arnone
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Marie Vallier
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University and Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Sébastien Hergalant
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Caroline Chabot
- CHRU-Nancy, Pediatric Hepato-Gastroenterology and Nutrition Unit, Department of Child Medicine and Clinical Genetics, Inserm U1256, Université de Lorraine, Nancy, France
| | - Ndeye Coumba Ndiaye
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - David Moulin
- IMoPA, UMR7365 CNRS-Université de Lorraine, CHRU de Nancy, Contrat d'interface, Nancy, France
| | | | - Jean-Marc Alberto
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Huguette Louis
- Department Inserm UMRS_1116 DCAC, Université de Lorraine, Nancy, France
- Cytometry Core Facility, UMS2008 IBSLor (CNRS-Université de Lorraine-INSERM), Campus Brabois-Santé, Nancy, France
| | - Olivier Boulard
- Laboratory of Cell Physiology, INSERM U1003, University of Lille, Lille, France
| | - Camille Mayeur
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Natacha Dreumont
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Kenneth Peuker
- Center for Regenerative Therapies, Technische Universität (TU) Dresden, Dresden, Germany
- Department of Medicine I, University Medical Center Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Anne Strigli
- Center for Regenerative Therapies, Technische Universität (TU) Dresden, Dresden, Germany
- Department of Medicine I, University Medical Center Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Sebastian Zeissig
- Center for Regenerative Therapies, Technische Universität (TU) Dresden, Dresden, Germany
- Department of Medicine I, University Medical Center Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Franck Hansmannel
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | | | - Tunay Kökten
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
| | - Laurent Peyrin-Biroulet
- Inserm U1256, Nutrition Genetics and Exposition NGERE, Université de Lorraine, Nancy, France
- Department of Gastroenterology, CHRU-Nancy, Université de Lorraine, Nancy, France
| |
Collapse
|
17
|
Chang MH, Levescot A, Nelson-Maney N, Blaustein RB, Winden KD, Morris A, Wactor A, Balu S, Grieshaber-Bouyer R, Wei K, Henderson LA, Iwakura Y, Clark RA, Rao DA, Fuhlbrigge RC, Nigrovic PA. Arthritis flares mediated by tissue-resident memory T cells in the joint. Cell Rep 2021; 37:109902. [PMID: 34706228 DOI: 10.1016/j.celrep.2021.109902] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 08/20/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022] Open
Abstract
Rheumatoid arthritis is a systemic autoimmune disease, but disease flares typically affect only a subset of joints, distributed in a distinctive pattern for each patient. Pursuing this intriguing pattern, we show that arthritis recurrence is mediated by long-lived synovial resident memory T cells (TRM). In three murine models, CD8+ cells bearing TRM markers remain in previously inflamed joints during remission. These cells are bona fide TRM, exhibiting a failure to migrate between joints, preferential uptake of fatty acids, and long-term residency. Disease flares result from TRM activation by antigen, leading to CCL5-mediated recruitment of circulating effector cells. Correspondingly, TRM depletion ameliorates recurrence in a site-specific manner. Human rheumatoid arthritis joint tissues contain a comparable CD8+-predominant TRM population, which is most evident in late-stage leukocyte-poor synovium, exhibiting limited T cell receptor diversity and a pro-inflammatory transcriptomic signature. Together, these findings establish synovial TRM as a targetable mediator of disease chronicity in autoimmune arthritis.
Collapse
Affiliation(s)
- Margaret H Chang
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anaïs Levescot
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Nathan Nelson-Maney
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Rachel B Blaustein
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kellen D Winden
- Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Allyn Morris
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alexandra Wactor
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Spoorthi Balu
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ricardo Grieshaber-Bouyer
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lauren A Henderson
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yoichiro Iwakura
- Center for Experimental Animal Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Rachael A Clark
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert C Fuhlbrigge
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Houser MC, Caudle WM, Chang J, Kannarkat GT, Yang Y, Kelly SD, Oliver D, Joers V, Shannon KM, Keshavarzian A, Tansey MG. Experimental colitis promotes sustained, sex-dependent, T-cell-associated neuroinflammation and parkinsonian neuropathology. Acta Neuropathol Commun 2021; 9:139. [PMID: 34412704 PMCID: PMC8375080 DOI: 10.1186/s40478-021-01240-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background The etiology of sporadic Parkinson’s disease (PD) remains uncertain, but genetic, epidemiological, and physiological overlap between PD and inflammatory bowel disease suggests that gut inflammation could promote dysfunction of dopamine-producing neurons in the brain. Mechanisms behind this pathological gut-brain effect and their interactions with sex and with environmental factors are not well understood but may represent targets for therapeutic intervention. Methods We sought to identify active inflammatory mechanisms which could potentially contribute to neuroinflammation and neurological disease in colon biopsies and peripheral blood immune cells from PD patients. Then, in mouse models, we assessed whether dextran sodium sulfate-mediated colitis could exert lingering effects on dopaminergic pathways in the brain and whether colitis increased vulnerability to a subsequent exposure to the dopaminergic neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We assessed the involvement of inflammatory mechanisms identified in the PD patients in colitis-related neurological dysfunction in male and female mice, utilizing mice lacking the Regulator of G-Protein Signaling 10 (RGS10)—an inhibitor of nuclear factor kappa B (NFκB)—to model enhanced NFκB activity, and mice in which CD8+ T-cells were depleted. Results High levels of inflammatory markers including CD8B and NFκB p65 were found in colon biopsies from PD patients, and reduced levels of RGS10 were found in immune cells in the blood. Male mice that experienced colitis exhibited sustained reductions in tyrosine hydroxylase but not in dopamine as well as sustained CD8+ T-cell infiltration and elevated Ifng expression in the brain. CD8+ T-cell depletion prevented colitis-associated reductions in dopaminergic markers in males. In both sexes, colitis potentiated the effects of MPTP. RGS10 deficiency increased baseline intestinal inflammation, colitis severity, and neuropathology. Conclusions This study identifies peripheral inflammatory mechanisms in PD patients and explores their potential to impact central dopaminergic pathways in mice. Our findings implicate a sex-specific interaction between gastrointestinal inflammation and neurologic vulnerability that could contribute to PD pathogenesis, and they establish the importance of CD8+ T-cells in this process in male mice. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s40478-021-01240-4.
Collapse
|
19
|
Barreto de Albuquerque J, Mueller C, Gungor B. Tissue-Resident T Cells in Chronic Relapsing-Remitting Intestinal Disorders. Cells 2021; 10:1882. [PMID: 34440651 PMCID: PMC8393248 DOI: 10.3390/cells10081882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 11/24/2022] Open
Abstract
Tissue-resident memory T (TRM) cells critically contribute to the rapid immunoprotection and efficient immunosurveillance against pathogens, particularly in barrier tissues, but also during anti-tumor responses. However, the involvement of TRM cells also in the induction and exacerbation of immunopathologies, notably in chronically relapsing auto-inflammatory disorders, is becoming increasingly recognized as a critical factor. Thus, TRM cells may also represent an attractive target in the management of chronic (auto-) inflammatory disorders, including multiple sclerosis, rheumatoid arthritis, celiac disease and inflammatory bowel diseases. In this review, we focus on current concepts of TRM cell biology, particularly in the intestine, and discuss recent findings on their involvement in chronic relapsing-remitting inflammatory disorders. Potential therapeutic strategies to interfere with these TRM cell-mediated immunopathologies are discussed.
Collapse
Affiliation(s)
| | | | - Bilgi Gungor
- Division of Experimental Pathology, Institute of Pathology, University of Bern, 3008 Bern, Switzerland;
| |
Collapse
|
20
|
Tkachev V, Kaminski J, Potter EL, Furlan SN, Yu A, Hunt DJ, McGuckin C, Zheng H, Colonna L, Gerdemann U, Carlson J, Hoffman M, Olvera J, English C, Baldessari A, Panoskaltsis-Mortari A, Watkins B, Qayed M, Suessmuth Y, Betz K, Bratrude B, Langston A, Horan JT, Ordovas-Montanes J, Shalek AK, Blazar BR, Roederer M, Kean LS. Spatiotemporal single-cell profiling reveals that invasive and tissue-resident memory donor CD8 + T cells drive gastrointestinal acute graft-versus-host disease. Sci Transl Med 2021; 13:13/576/eabc0227. [PMID: 33441422 DOI: 10.1126/scitranslmed.abc0227] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022]
Abstract
Organ infiltration by donor T cells is critical to the development of acute graft-versus-host disease (aGVHD) in recipients after allogeneic hematopoietic stem cell transplant (allo-HCT). However, deconvoluting the transcriptional programs of newly recruited donor T cells from those of tissue-resident T cells in aGVHD target organs remains a challenge. Here, we combined the serial intravascular staining technique with single-cell RNA sequencing to dissect the tightly connected processes by which donor T cells initially infiltrate tissues and then establish a pathogenic tissue residency program in a rhesus macaque allo-HCT model that develops aGVHD. Our results enabled creation of a spatiotemporal map of the transcriptional programs controlling donor CD8+ T cell infiltration into the primary aGVHD target organ, the gastrointestinal (GI) tract. We identified the large and small intestines as the only two sites demonstrating allo-specific, rather than lymphodepletion-driven, T cell infiltration. GI-infiltrating donor CD8+ T cells demonstrated a highly activated, cytotoxic phenotype while simultaneously developing a canonical tissue-resident memory T cell (TRM) transcriptional signature driven by interleukin-15 (IL-15)/IL-21 signaling. We found expression of a cluster of genes directly associated with tissue invasiveness, including those encoding adhesion molecules (ITGB2), specific chemokines (CCL3 and CCL4L1) and chemokine receptors (CD74), as well as multiple cytoskeletal proteins. This tissue invasion transcriptional signature was validated by its ability to discriminate the CD8+ T cell transcriptome of patients with GI aGVHD from those of GVHD-free patients. These results provide insights into the mechanisms controlling tissue occupancy of target organs by pathogenic donor CD8+ TRM cells during aGVHD in primate transplant recipients.
Collapse
Affiliation(s)
- Victor Tkachev
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - James Kaminski
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - E Lake Potter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20858, USA
| | - Scott N Furlan
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Alison Yu
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel J Hunt
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Connor McGuckin
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hengqi Zheng
- University of Washington, Seattle, WA 98195, USA
| | - Lucrezia Colonna
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Ulrike Gerdemann
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Michelle Hoffman
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Joe Olvera
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Chris English
- Washington National Primate Research Center, Seattle, WA 98195, USA
| | | | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55454, USA
| | | | - Muna Qayed
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Kayla Betz
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Brandi Bratrude
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - John T Horan
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jose Ordovas-Montanes
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Division of Gastroenterology, Boston Children's Hospital and Program in Immunology, Harvard Medical School, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Alex K Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55454, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20858, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Huang D, Chen X, Zeng X, Lao L, Li J, Xing Y, Lu Y, Ouyang Q, Chen J, Yang L, Su F, Yao H, Liu Q, Su S, Song E. Targeting regulator of G protein signaling 1 in tumor-specific T cells enhances their trafficking to breast cancer. Nat Immunol 2021; 22:865-879. [PMID: 34140678 DOI: 10.1038/s41590-021-00939-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 04/22/2021] [Indexed: 02/05/2023]
Abstract
Reduced infiltration of anti-tumor lymphocytes remains a major cause of tumor immune evasion and is correlated with poor cancer survival. Here, we found that upregulation of regulator of G protein signaling (RGS)1 in helper TH1 cells and cytotoxic T lymphocytes (CTLs) reduced their trafficking to and survival in tumors and was associated with shorter survival of patients with breast and lung cancer. RGS1 was upregulated by type II interferon (IFN)-signal transducer and activator of transcription (STAT)1 signaling and impaired trafficking of circulating T cells to tumors by inhibiting calcium influx and suppressing activation of the kinases ERK and AKT. RGS1 knockdown in adoptively transferred tumor-specific CTLs significantly increased their infiltration and survival in breast and lung tumor grafts and effectively inhibited tumor growth in vivo, which was further improved when combined with programmed death ligand (PD-L)1 checkpoint inhibition. Our findings reveal RGS1 is important for tumor immune evasion and suggest that targeting RGS1 may provide a new strategy for tumor immunotherapy.
Collapse
MESH Headings
- Animals
- Apoptosis
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/therapy
- Cell Line, Tumor
- Chemokines/metabolism
- Chemotaxis, Leukocyte
- Coculture Techniques
- Cytotoxicity, Immunologic
- Female
- Humans
- Immunotherapy, Adoptive
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/transplantation
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Microscopy, Fluorescence
- Microscopy, Video
- RGS Proteins/genetics
- RGS Proteins/metabolism
- Signal Transduction
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Time Factors
- Time-Lapse Imaging
- Tumor Cells, Cultured
- Tumor Escape
- Mice
Collapse
Affiliation(s)
- Di Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Xueman Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Xin Zeng
- Bioland Laboratory, Guangzhou, China
- Program of Molecular Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Liyan Lao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Jiaqian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yiwen Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Qian Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Jianing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Linbin Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Fengxi Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Bioland Laboratory, Guangzhou, China.
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Bioland Laboratory, Guangzhou, China.
- Program of Molecular Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
- Fountain-Valley Institute for Life Sciences, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| |
Collapse
|
22
|
McFarland AP, Yalin A, Wang SY, Cortez VS, Landsberger T, Sudan R, Peng V, Miller HL, Ricci B, David E, Faccio R, Amit I, Colonna M. Multi-tissue single-cell analysis deconstructs the complex programs of mouse natural killer and type 1 innate lymphoid cells in tissues and circulation. Immunity 2021; 54:1320-1337.e4. [PMID: 33945787 PMCID: PMC8312473 DOI: 10.1016/j.immuni.2021.03.024] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 01/28/2021] [Accepted: 03/30/2021] [Indexed: 12/18/2022]
Abstract
Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1s) are heterogenous innate lymphocytes broadly defined in mice as Lin-NK1.1+NKp46+ cells that express the transcription factor T-BET and produce interferon-γ. The ILC1 definition primarily stems from studies on liver and small intestinal populations. However, NK1.1+NKp46+ cells in the salivary glands, uterus, adipose, and other tissues exhibit nonuniform programs that differ from those of liver or intestinal ILC1s or NK cells. Here, we performed single-cell RNA sequencing on murine NK1.1+NKp46+ cells from blood, spleen, various tissues, and solid tumors. We identified gene expression programs of tissue-specific ILC1s, tissue-specific NK cells, and non-tissue-specific populations in blood, spleen, and other tissues largely corresponding to circulating cells. Moreover, we found that circulating NK cell programs were reshaped in tumor-bearing mice. Core programs of circulating and tumor NK cells paralleled conserved human NK cells signatures, advancing our understanding of the human NK-ILC1 spectrum.
Collapse
Affiliation(s)
- Adelle P McFarland
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Adam Yalin
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shuang-Yin Wang
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Victor S Cortez
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tomer Landsberger
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Raki Sudan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vincent Peng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah L Miller
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Biancamaria Ricci
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Roberta Faccio
- Department of Orthopedics, Washington University School of Medicine, St. Louis, MO, USA; Shriners Children's Hospital in St. Louis, St. Louis, MO, USA
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
23
|
Kaufmann M, Evans H, Schaupp AL, Engler JB, Kaur G, Willing A, Kursawe N, Schubert C, Attfield KE, Fugger L, Friese MA. Identifying CNS-colonizing T cells as potential therapeutic targets to prevent progression of multiple sclerosis. MED 2021; 2:296-312.e8. [PMID: 33748804 PMCID: PMC7966680 DOI: 10.1016/j.medj.2021.01.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/17/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Multiple sclerosis (MS), an autoimmune disease of the central nervous system (CNS), can be suppressed in its early stages but eventually becomes clinically progressive and unresponsive to therapy. Here, we investigate whether the therapeutic resistance of progressive MS can be attributed to chronic immune cell accumulation behind the blood-brain barrier (BBB). METHODS We systematically track CNS-homing immune cells in the peripheral blood of 31 MS patients and 31 matched healthy individuals in an integrated analysis of 497,705 single-cell transcriptomes and 355,433 surface protein profiles from 71 samples. Through spatial RNA sequencing, we localize these cells in post mortem brain tissue of 6 progressive MS patients contrasted against 4 control brains (20 samples, 85,000 spot transcriptomes). FINDINGS We identify a specific pathogenic CD161+/lymphotoxin beta (LTB)+ T cell population that resides in brains of progressive MS patients. Intriguingly, our data suggest that the colonization of the CNS by these T cells may begin earlier in the disease course, as they can be mobilized to the blood by usage of the integrin-blocking antibody natalizumab in relapsing-remitting MS patients. CONCLUSIONS As a consequence, we lay the groundwork for a therapeutic strategy to deplete CNS-homing T cells before they can fuel treatment-resistant progression. FUNDING This study was supported by funding from the University Medical Center Hamburg-Eppendorf, the Stifterverband für die Deutsche Wissenschaft, the OAK Foundation, Medical Research Council UK, and Wellcome.
Collapse
Affiliation(s)
- Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Hayley Evans
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Anna-Lena Schaupp
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gurman Kaur
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Anne Willing
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Kursawe
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Charlotte Schubert
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrine E. Attfield
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Manuel A. Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
24
|
Shannon MJ, Mace EM. Natural Killer Cell Integrins and Their Functions in Tissue Residency. Front Immunol 2021; 12:647358. [PMID: 33777044 PMCID: PMC7987804 DOI: 10.3389/fimmu.2021.647358] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Integrins are transmembrane receptors associated with adhesion and migration and are often highly differentially expressed receptors amongst natural killer cell subsets in microenvironments. Tissue resident natural killer cells are frequently defined by their differential integrin expression compared to other NK cell subsets, and integrins can further localize tissue resident NK cells to tissue microenvironments. As such, integrins play important roles in both the phenotypic and functional identity of NK cell subsets. Here we review the expression of integrin subtypes on NK cells and NK cell subsets with the goal of better understanding how integrin selection can dictate tissue residency and mediate function from the nanoscale to the tissue environment.
Collapse
Affiliation(s)
| | - Emily M. Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
25
|
Stein JV, Ruef N, Wissmann S. Organ-Specific Surveillance and Long-Term Residency Strategies Adapted by Tissue-Resident Memory CD8 + T Cells. Front Immunol 2021; 12:626019. [PMID: 33659008 PMCID: PMC7917134 DOI: 10.3389/fimmu.2021.626019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/26/2021] [Indexed: 01/27/2023] Open
Abstract
Tissue-resident CD8+ T cells (CD8+ TRM) populate lymphoid and non-lymphoid tissues after infections as first line of defense against re-emerging pathogens. To achieve host protection, CD8+ TRM have developed surveillance strategies that combine dynamic interrogation of pMHC complexes on local stromal and hematopoietic cells with long-term residency. Factors mediating CD8+ TRM residency include CD69, a surface receptor opposing the egress-promoting S1P1, CD49a, a collagen-binding integrin, and CD103, which binds E-cadherin on epithelial cells. Moreover, the topography of the tissues of residency may influence TRM retention and surveillance strategies. Here, we provide a brief summary of these factors to examine how CD8+ TRM reconcile constant migratory behavior with their long-term commitment to local microenvironments, with a focus on epithelial barrier organs and exocrine glands with mixed connective-epithelial tissue composition.
Collapse
Affiliation(s)
- Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Stefanie Wissmann
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
26
|
Montaldo P, Cunnington A, Oliveira V, Swamy R, Bandya P, Pant S, Lally PJ, Ivain P, Mendoza J, Atreja G, Padmesh V, Baburaj M, Sebastian M, Yasashwi I, Kamalarathnam C, Chandramohan R, Mangalabharathi S, Kumaraswami K, Kumar S, Benakappa N, Manerkar S, Mondhkar J, Prakash V, Sajjid M, Seeralar A, Jahan I, Moni SC, Shahidullah M, Sujatha R, Chandrasekaran M, Ramji S, Shankaran S, Kaforou M, Herberg J, Thayyil S. Transcriptomic profile of adverse neurodevelopmental outcomes after neonatal encephalopathy. Sci Rep 2020; 10:13100. [PMID: 32753750 PMCID: PMC7403382 DOI: 10.1038/s41598-020-70131-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 06/16/2020] [Indexed: 12/20/2022] Open
Abstract
A rapid and early diagnostic test to identify the encephalopathic babies at risk of adverse outcome may accelerate the development of neuroprotectants. We examined if a whole blood transcriptomic signature measured soon after birth, predicts adverse neurodevelopmental outcome eighteen months after neonatal encephalopathy. We performed next generation sequencing on whole blood ribonucleic acid obtained within six hours of birth from the first 47 encephalopathic babies recruited to the Hypothermia for Encephalopathy in Low and middle-income countries (HELIX) trial. Two infants with blood culture positive sepsis were excluded, and the data from remaining 45 were analysed. A total of 855 genes were significantly differentially expressed between the good and adverse outcome groups, of which RGS1 and SMC4 were the most significant. Biological pathway analysis adjusted for gender, trial randomisation allocation (cooling therapy versus usual care) and estimated blood leukocyte proportions revealed over-representation of genes from pathways related to melatonin and polo-like kinase in babies with adverse outcome. These preliminary data suggest that transcriptomic profiling may be a promising tool for rapid risk stratification in neonatal encephalopathy. It may provide insights into biological mechanisms and identify novel therapeutic targets for neuroprotection.
Collapse
Affiliation(s)
- Paolo Montaldo
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK. .,Neonatal Unit, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| | - Aubrey Cunnington
- Paediatric Infectious Diseases, Department of Infectious Diseases, Imperial College London, London, UK
| | - Vania Oliveira
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Ravi Swamy
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Prathik Bandya
- Neonatal Medicine, Indira Gandhi Institute of Child Health, Bangalore, Karnataka, India
| | - Stuti Pant
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Peter J Lally
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Phoebe Ivain
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Josephine Mendoza
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Gaurav Atreja
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Vadakepat Padmesh
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Mythili Baburaj
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Monica Sebastian
- Neonatal Medicine, Institute of Child Health, Madras Medical College, Tamil Nadu, Chennai, India
| | - Indiramma Yasashwi
- Neonatal Medicine, Indira Gandhi Institute of Child Health, Bangalore, Karnataka, India
| | - Chinnathambi Kamalarathnam
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Rema Chandramohan
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Sundaram Mangalabharathi
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Kumutha Kumaraswami
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Shobha Kumar
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Naveen Benakappa
- Neonatal Medicine, Indira Gandhi Institute of Child Health, Bangalore, Karnataka, India
| | | | | | - Vinayagam Prakash
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Mohammed Sajjid
- Neonatal Medicine, Institute of Obstetrics and Gynaecology, Madras Medical College, Chennai, Tamil Nadu, India
| | - Arasar Seeralar
- Neonatal Medicine, Institute of Child Health, Madras Medical College, Tamil Nadu, Chennai, India
| | - Ismat Jahan
- Neonatal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | | | - Mohammod Shahidullah
- Neonatal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Radhika Sujatha
- Neonatal Medicine, Government Medical College, Thiruvananthapuram, Kerala, India
| | - Manigandan Chandrasekaran
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| | - Siddarth Ramji
- Neonatal Medicine, Maulana Azad Medical College, New Delhi, Delhi, India
| | - Seetha Shankaran
- Neonatal-Perinatal Medicine, Wayne State University, Detroit, MI, USA
| | - Myrsini Kaforou
- Paediatric Infectious Diseases, Department of Infectious Diseases, Imperial College London, London, UK
| | - Jethro Herberg
- Paediatric Infectious Diseases, Department of Infectious Diseases, Imperial College London, London, UK
| | - Sudhin Thayyil
- Department of Brain Sciences, Centre for Perinatal Neuroscience, Imperial College London, London, UK
| |
Collapse
|
27
|
Lämmermann T, Kastenmüller W. Concepts of GPCR-controlled navigation in the immune system. Immunol Rev 2020; 289:205-231. [PMID: 30977203 PMCID: PMC6487968 DOI: 10.1111/imr.12752] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/01/2019] [Accepted: 02/03/2019] [Indexed: 12/11/2022]
Abstract
G‐protein–coupled receptor (GPCR) signaling is essential for the spatiotemporal control of leukocyte dynamics during immune responses. For efficient navigation through mammalian tissues, most leukocyte types express more than one GPCR on their surface and sense a wide range of chemokines and chemoattractants, leading to basic forms of leukocyte movement (chemokinesis, haptokinesis, chemotaxis, haptotaxis, and chemorepulsion). How leukocytes integrate multiple GPCR signals and make directional decisions in lymphoid and inflamed tissues is still subject of intense research. Many of our concepts on GPCR‐controlled leukocyte navigation in the presence of multiple GPCR signals derive from in vitro chemotaxis studies and lower vertebrates. In this review, we refer to these concepts and critically contemplate their relevance for the directional movement of several leukocyte subsets (neutrophils, T cells, and dendritic cells) in the complexity of mouse tissues. We discuss how leukocyte navigation can be regulated at the level of only a single GPCR (surface expression, competitive antagonism, oligomerization, homologous desensitization, and receptor internalization) or multiple GPCRs (synergy, hierarchical and non‐hierarchical competition, sequential signaling, heterologous desensitization, and agonist scavenging). In particular, we will highlight recent advances in understanding GPCR‐controlled leukocyte navigation by intravital microscopy of immune cells in mice.
Collapse
Affiliation(s)
- Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | |
Collapse
|
28
|
Stein JV, Ruef N. Regulation of global CD8 + T-cell positioning by the actomyosin cytoskeleton. Immunol Rev 2020; 289:232-249. [PMID: 30977193 DOI: 10.1111/imr.12759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 12/12/2022]
Abstract
CD8+ T cells have evolved as one of the most motile mammalian cell types, designed to continuously scan peptide-major histocompatibility complexes class I on the surfaces of other cells. Chemoattractants and adhesion molecules direct CD8+ T-cell homing to and migration within secondary lymphoid organs, where these cells colocalize with antigen-presenting dendritic cells in confined tissue volumes. CD8+ T-cell activation induces a switch to infiltration of non-lymphoid tissue (NLT), which differ in their topology and biophysical properties from lymphoid tissue. Here, we provide a short overview on regulation of organism-wide trafficking patterns during naive T-cell recirculation and their switch to non-lymphoid tissue homing during activation. The migratory lifestyle of CD8+ T cells is regulated by their actomyosin cytoskeleton, which translates chemical signals from surface receptors into mechanical work. We explore how properties of the actomyosin cytoskeleton and its regulators affect CD8+ T cell function in lymphoid and non-lymphoid tissue, combining recent findings in the field of cell migration and actin network regulation with tissue anatomy. Finally, we hypothesize that under certain conditions, intrinsic regulation of actomyosin dynamics may render NLT CD8+ T-cell populations less dependent on input from extrinsic signals during tissue scanning.
Collapse
Affiliation(s)
- Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
29
|
Singh RS, Singh A, Batra G, Kaur H, Medhi B. Novel targets for drug discovery in celiac disease. Indian J Pharmacol 2019; 51:359-365. [PMID: 31831931 PMCID: PMC6892008 DOI: 10.4103/ijp.ijp_679_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 11/21/2022] Open
Abstract
Celiac disease is a lifelong, immunological disorder induced by dietary protein-gluten, in a genetically susceptible populations, resulting in different clinical manifestations, the release of antibodies, and damage to the intestinal mucosa. The only recommended therapy for the disease is to strictly follow a gluten-free diet (GFD), which is difficult to comply with. A GFD is found to be ineffective in some active Celiac disease cases. Therefore, there is an unmet need for an alternative nondietary therapeutic approach. The review focuses on the novel drug targets for Celiac disease.
Collapse
Affiliation(s)
- Rahul Soloman Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashutosh Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gitika Batra
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Hardeep Kaur
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
30
|
Li H, Kaiser TK, Borschiwer M, Bohnenberger H, Reichardt SD, Lühder F, Walter L, Dressel R, Meijsing SH, Reichardt HM. Glucocorticoid resistance of allogeneic T cells alters the gene expression profile in the inflamed small intestine of mice suffering from acute graft-versus-host disease. J Steroid Biochem Mol Biol 2019; 195:105485. [PMID: 31561002 DOI: 10.1016/j.jsbmb.2019.105485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/20/2019] [Accepted: 09/21/2019] [Indexed: 01/20/2023]
Abstract
Glucocorticoids (GCs) play an important role in controlling acute graft-versus-host disease (aGvHD), a frequent complication of allogeneic hematopoietic stem cell transplantation. The anti-inflammatory activity of GCs is mainly ascribed to the modulation of T cells and macrophages, for which reason a genetically induced GC resistance of either of these cell types causes aggravated aGvHD. Since only a few genes are currently known that are differentially regulated under these conditions, we analyzed the expression of 54 candidate genes in the inflamed small intestine of mice suffering from aGvHD when either allogeneic T cells or host myeloid cells were GC resistant using a microfluidic dynamic array platform for high-throughput quantitative PCR. The majority of genes categorized as cytokines (e.g. Il2, Il6), chemokines (e.g. Ccl2, Cxcl1), cell surface receptors (e.g. Fasl, Ctla4) and intracellular molecules (e.g. Dusp1, Arg1) were upregulated in mice transplanted with GC resistant allogeneic T cells. Moreover, the expression of several genes linked to energy metabolism (e.g. Glut1) was altered. Surprisingly, mice harboring GC resistant myeloid cells showed almost no changes in gene expression despite their fatal disease course after aGvHD induction. To identify additional genes in the inflamed small intestine that were affected by a GC resistance of allogeneic T cells, we performed an RNAseq analysis, which uncovered more than 500 differentially expressed transcripts (e.g. Cxcr6, Glut3, Otc, Aoc1, Il1r1, Sphk1) that were enriched for biological processes associated with inflammation and tissue disassembly. The changes in gene expression could be confirmed during full-blown disease but hardly any of them in the preclinical phase using high-throughput quantitative PCR. Further analysis of some of these genes revealed a highly selective expression pattern in T cells, intestinal epithelial cells and macrophages, which correlated with their regulation during disease progression. Collectively, we identified an altered gene expression profile caused by GC resistance of transplanted allogeneic T cells, which could help to define new targets for aGvHD therapy.
Collapse
Affiliation(s)
- Hu Li
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany
| | - Tina K Kaiser
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany
| | - Marina Borschiwer
- Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195 Berlin, Germany
| | - Hanibal Bohnenberger
- University Medical Center Göttingen, Institute for Pathology, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Sybille D Reichardt
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany
| | - Fred Lühder
- University Medical Center Göttingen, Institute for Neuroimmunology and Multiple Sclerosis Research, von-Siebold-Straße 3a, 37075 Göttingen, Germany
| | - Lutz Walter
- German Primate Center, Leibniz Institute for Primate Research, Primate Genetics Laboratory, Kellnerweg 4, 37077 Göttingen, Germany
| | - Ralf Dressel
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany
| | | | - Holger M Reichardt
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany.
| |
Collapse
|
31
|
Pattacini L, Woodward Davis A, Czartoski J, Mair F, Presnell S, Hughes SM, Hyrien O, Lentz GM, Kirby AC, Fialkow MF, Hladik F, Prlic M, Lund JM. A pro-inflammatory CD8+ T-cell subset patrols the cervicovaginal tract. Mucosal Immunol 2019; 12:1118-1129. [PMID: 31312028 PMCID: PMC6717561 DOI: 10.1038/s41385-019-0186-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 02/04/2023]
Abstract
The immune system of the cervicovaginal tract (CVT) must balance immunosurveillance and active immunity against pathogens with maintenance of tolerance to resident microbiota and to fetal and partner antigens for reproductive purposes. Thus, we predicted that CVT immunity is characterized by distinctive features compared to blood and other tissue compartments. Indeed, we found that CVT CD8+ T-cells had unique transcriptional profiles, particularly in their cytokine signature, compared to that reported for CD8+ T-cells in other tissue sites. Among these CVT CD8+ T-cells, we identified a CD69- CD103- subset that was characterized by reduced migration in response to tissue-exit signals and higher pro-inflammatory potential as compared to their blood counterpart. These inflammatory mucosal CD8+ T-cells (Tim) were increased in frequency in the CVT of individuals with chronic infection, pointing to a potential role in perpetuating inflammation. Our findings highlight the specialized nature of immunity within the CVT and identify Tim cells as potential therapeutic targets to tame tissue inflammation upon chronic infection.
Collapse
Affiliation(s)
- Laura Pattacini
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Amanda Woodward Davis
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Julie Czartoski
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Florian Mair
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Scott Presnell
- System Immunology Division, Benaroya Research Institute, Seattle, WA, U.S.A
| | - Sean M. Hughes
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Gretchen M. Lentz
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, U.S.A
| | - Anna C. Kirby
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, U.S.A
| | - Michael F. Fialkow
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, U.S.A
| | - Florian Hladik
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, U.S.A
| | - Martin Prlic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Jennifer M. Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A.,Department of Global Health, University of Washington, Seattle, WA, U.S.A
| |
Collapse
|
32
|
Dotan I, Allez M, Danese S, Keir M, Tole S, McBride J. The role of integrins in the pathogenesis of inflammatory bowel disease: Approved and investigational anti-integrin therapies. Med Res Rev 2019; 40:245-262. [PMID: 31215680 PMCID: PMC6973243 DOI: 10.1002/med.21601] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/12/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease (IBD) is characterized by uncontrolled inflammation in the gastrointestinal tract. The underlying pathobiology of IBD includes an increase in infiltrating gut-homing lymphocytes. Although lymphocyte homing is typically a tightly regulated and stepwise process involving multiple integrins and adhesion molecules expressed on endothelial cells, the distinct roles of integrin-expressing immune cells is not fully understood in the pathology of IBD. In this review, we detail the involvement of integrins expressed on specific lymphocyte subsets in the pathogenesis of IBD and discuss the current status of approved and investigational integrin-targeted therapies.
Collapse
Affiliation(s)
- Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matthieu Allez
- Department of Gastroenterology, Hôpital Saint-Louis, AP-HP, INSERM U1160, University Denis Diderot, Paris, France
| | - Silvio Danese
- Gastrointestinal Immunopathology Laboratory and IBD Unit, Humanitas Clinical and Research Center, Milan, Italy
| | - Mary Keir
- Department of Research and Early Development, Genentech, South San Francisco, California
| | - Swati Tole
- Department of Product Development, Genentech, South San Francisco, California
| | - Jacqueline McBride
- Department of Research and Early Development, Genentech, South San Francisco, California
| |
Collapse
|
33
|
Hu X, Tang J, Zeng G, Hu X, Bao P, Wu J, Liang Y, Deng W, Tang Y. RGS1 silencing inhibits the inflammatory response and angiogenesis in rheumatoid arthritis rats through the inactivation of Toll‐like receptor signaling pathway. J Cell Physiol 2019; 234:20432-20442. [PMID: 31012109 DOI: 10.1002/jcp.28645] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Xumin Hu
- Department of Orthopedics, Sun Yat‐Sen Memorial Hospital Sun Yat‐Sen University Guangzhou P.R. China
| | - Jianhua Tang
- Department of Orthopedics The People's Hospital of Gaozhou Gaozhou P.R. China
| | - Gang Zeng
- Department of Orthopedics, Sun Yat‐Sen Memorial Hospital Sun Yat‐Sen University Guangzhou P.R. China
| | - Xuyun Hu
- Center for Medical Genetics, Beijing Children's Hospital Capital Medical University, National Center for Children's Health Beijing P.R. China
| | - Peng Bao
- Medical Department of Hospital of Stomatology Sun Yat‐Sen University Guangzhou P.R. China
| | - Jionglin Wu
- Department of Orthopedics, Sun Yat‐Sen Memorial Hospital Sun Yat‐Sen University Guangzhou P.R. China
| | - Yuwei Liang
- Department of Orthopedics, Sun Yat‐Sen Memorial Hospital Sun Yat‐Sen University Guangzhou P.R. China
| | - Weixi Deng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat‐Sen Memorial Hospital Sun Yat‐Sen University Guangzhou P.R. China
| | - Yong Tang
- Department of Orthopedics, Sun Yat‐Sen Memorial Hospital Sun Yat‐Sen University Guangzhou P.R. China
| |
Collapse
|
34
|
Squires KE, Montañez-Miranda C, Pandya RR, Torres MP, Hepler JR. Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Disease. Pharmacol Rev 2018; 70:446-474. [PMID: 29871944 DOI: 10.1124/pr.117.015354] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulators of G protein signaling (RGS) proteins modulate the physiologic actions of many neurotransmitters, hormones, and other signaling molecules. Human RGS proteins comprise a family of 20 canonical proteins that bind directly to G protein-coupled receptors/G protein complexes to limit the lifetime of their signaling events, which regulate all aspects of cell and organ physiology. Genetic variations account for diverse human traits and individual predispositions to disease. RGS proteins contribute to many complex polygenic human traits and pathologies such as hypertension, atherosclerosis, schizophrenia, depression, addiction, cancers, and many others. Recent analysis indicates that most human diseases are due to extremely rare genetic variants. In this study, we summarize physiologic roles for RGS proteins and links to human diseases/traits and report rare variants found within each human RGS protein exome sequence derived from global population studies. Each RGS sequence is analyzed using recently described bioinformatics and proteomic tools for measures of missense tolerance ratio paired with combined annotation-dependent depletion scores, and protein post-translational modification (PTM) alignment cluster analysis. We highlight selected variants within the well-studied RGS domain that likely disrupt RGS protein functions and provide comprehensive variant and PTM data for each RGS protein for future study. We propose that rare variants in functionally sensitive regions of RGS proteins confer profound change-of-function phenotypes that may contribute, in newly appreciated ways, to complex human diseases and/or traits. This information provides investigators with a valuable database to explore variation in RGS protein function, and for targeting RGS proteins as future therapeutic targets.
Collapse
Affiliation(s)
- Katherine E Squires
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Carolina Montañez-Miranda
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Rushika R Pandya
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Matthew P Torres
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - John R Hepler
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| |
Collapse
|
35
|
Human Tissue-Resident Memory T Cells Are Defined by Core Transcriptional and Functional Signatures in Lymphoid and Mucosal Sites. Cell Rep 2018; 20:2921-2934. [PMID: 28930685 DOI: 10.1016/j.celrep.2017.08.078] [Citation(s) in RCA: 711] [Impact Index Per Article: 118.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/03/2017] [Accepted: 08/23/2017] [Indexed: 01/18/2023] Open
Abstract
Tissue-resident memory T cells (TRMs) in mice mediate optimal protective immunity to infection and vaccination, while in humans, the existence and properties of TRMs remain unclear. Here, we use a unique human tissue resource to determine whether human tissue memory T cells constitute a distinct subset in diverse mucosal and lymphoid tissues. We identify a core transcriptional profile within the CD69+ subset of memory CD4+ and CD8+ T cells in lung and spleen that is distinct from that of CD69- TEM cells in tissues and circulation and defines human TRMs based on homology to the transcriptional profile of mouse CD8+ TRMs. Human TRMs in diverse sites exhibit increased expression of adhesion and inhibitory molecules, produce both pro-inflammatory and regulatory cytokines, and have reduced turnover compared with circulating TEM, suggesting unique adaptations for in situ immunity. Together, our results provide a unifying signature for human TRM and a blueprint for designing tissue-targeted immunotherapies.
Collapse
|
36
|
Patel J, Chuaiphichai S, Douglas G, Gorvin CM, Channon KM. Vascular wall regulator of G-protein signalling-1 (RGS-1) is required for angiotensin II-mediated blood pressure control. Vascul Pharmacol 2018; 108:15-22. [PMID: 29654907 PMCID: PMC6073721 DOI: 10.1016/j.vph.2018.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 03/18/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
Abstract
G-Protein coupled receptors (GPCRs) activate intracellular signalling pathways by coupling to heterotrimeric G-proteins that control many physiological processes including blood pressure homeostasis. The Regulator of G-Protein Signalling-1 (RGS1) controls the magnitude and duration of downstream GPCR signalling by acting as a GTPase-activating protein for specific Gα-proteins. RGS1 has contrasting roles in haematopoietic and non-haematopoietic cells. Rgs1−/−ApoE−/− mice are protected from Angiotensin II (Ang II)-induced aortic aneurysm rupture. Conversely, Ang II treatment increases systolic blood pressure to a greater extent in Rgs1−/−ApoE−/− mice than ApoE−/− mice, independent of its role in myeloid cells. However the precise role of RGS1 in hypertension and vascular-derived cells remains unknown. We determined the effects of Rgs1 deletion on vascular function in ApoE−/− mice. Rgs1 deletion led to enhanced vasoconstriction in aortas and mesenteric arteries from ApoE−/− mice in response to phenylephrine (PE) and U46619 respectively. Rgs1 was shown to have a role in the vasculature, with endothelium-dependent vasodilation being impaired, and endothelium-independent dilatation to SNP being enhanced in Rgs1−/−ApoE−/− mesenteric arteries. To address the downstream signalling pathways in vascular smooth muscle cells (VSMCs) in response to Ang II-stimulation, we assessed pErk1/2, pJNK and pp38 MAPK activation in VSMCs transiently transfected with Rgs1. pErk1/2 signalling but not pJNK and pp38 signalling was impaired in the presence of Rgs1. Furthermore, we demonstrated that the enhanced contractile response to PE in Rgs1−/−ApoE−/− aortas was reduced by a MAPK/Erk (MEK) inhibitor and an L-type voltage gated calcium channel antagonist, suggesting that Erk1/2 signalling and calcium influx are major effectors of Rgs1-mediated vascular contractile responses, respectively. These findings indicate RGS1 is a novel regulator of blood pressure homeostasis and highlight RGS1-controlled signalling pathways in the vasculature that may be new drug development targets for hypertension.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/physiopathology
- Blood Pressure/genetics
- Calcium Signaling
- Cell Line
- Disease Models, Animal
- Hypertension/chemically induced
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Male
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/physiopathology
- Mice, Knockout, ApoE
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Phosphorylation
- RGS Proteins/deficiency
- RGS Proteins/genetics
- RGS Proteins/metabolism
- Receptor, Angiotensin, Type 1/metabolism
- Vasoconstriction
- Vasodilation
Collapse
Affiliation(s)
- Jyoti Patel
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK.
| | - Surawee Chuaiphichai
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Caroline M Gorvin
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| |
Collapse
|
37
|
Wang D. The essential role of G protein-coupled receptor (GPCR) signaling in regulating T cell immunity. Immunopharmacol Immunotoxicol 2018; 40:187-192. [PMID: 29433403 DOI: 10.1080/08923973.2018.1434792] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
AIM The aim of this paper is to clarify the critical role of GPCR signaling in T cell immunity. METHODS The G protein-coupled receptors (GPCRs) are the most common targets in current pharmaceutical industry, and represent the largest and most versatile family of cell surface communicating molecules. GPCRs can be activated by a diverse array of ligands including neurotransmitters, chemokines as well as sensory stimuli. Therefore, GPCRs are involved in many key cellular and physiological processes, such as sense of light, taste and smell, neurotransmission, metabolism, endocrine and exocrine secretion. In recent years, GPCRs have been found to play an important role in immune system. T cell is an important type of immune cell, which plays a central role in cell-mediated immunity. A variety of GPCRs and their signaling mediators (RGS proteins, GRKs and β-arrestin) have been found to express in T cells and involved T cell-mediated immunity. We will summarize the role of GPCR signaling and their regulatory molecules in T cell activation, homeostasis and function in this article. RESULTS GPCR signaling plays an important role in T cell activation, homeostasis and function. CONCLUSION GPCR signaling is critical in regulating T cell immunity.
Collapse
Affiliation(s)
- Dashan Wang
- a Molecular Biology Research Center, Key Medical Health Laboratory for Laboratory Medicine of Shandong Province, Department of Laboratory Medicine , Shandong Medical College , Linyi , Shandong , China
| |
Collapse
|
38
|
ATF3 negatively regulates cellular antiviral signaling and autophagy in the absence of type I interferons. Sci Rep 2017; 7:8789. [PMID: 28821775 PMCID: PMC5562757 DOI: 10.1038/s41598-017-08584-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/21/2017] [Indexed: 01/19/2023] Open
Abstract
Stringent regulation of antiviral signaling and cellular autophagy is critical for the host response to virus infection. However, little is known how these cellular processes are regulated in the absence of type I interferon signaling. Here, we show that ATF3 is induced following Japanese encephalitis virus (JEV) infection, and regulates cellular antiviral and autophagy pathways in the absence of type I interferons in mouse neuronal cells. We have identified new targets of ATF3 and show that it binds to the promoter regions of Stat1, Irf9, Isg15 and Atg5 thereby inhibiting cellular antiviral signaling and autophagy. Consistent with these observations, ATF3-depleted cells showed enhanced antiviral responses and induction of robust autophagy. Furthermore, we show that JEV replication was significantly reduced in ATF3-depleted cells. Our findings identify ATF3 as a negative regulator of antiviral signaling and cellular autophagy in mammalian cells, and demonstrate its important role in JEV life cycle.
Collapse
|
39
|
Banaganapalli B, Rashidi O, Saadah OI, Wang J, Khan IA, Al-Aama JY, Shaik NA, Elango R. Comprehensive Computational Analysis of GWAS Loci Identifies CCR2 as a Candidate Gene for Celiac Disease Pathogenesis. J Cell Biochem 2017; 118:2193-2207. [PMID: 28059456 DOI: 10.1002/jcb.25864] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/04/2017] [Indexed: 01/01/2023]
Abstract
Celiac disease (CD) is a gluten intolerance disorder with known genetic contribution. The recent fine mapping and genome-wide association studies (GWAS) have identified up to 57 non-HLA CD susceptibility SNPs, majority of which are non-coding variants lacking any functional annotation. Therefore, we adopted multidimensional computational approach for uncovering the plausible mechanisms through which these GWAS SNPs are connected to CD pathogenesis. At initial phase, we identified that 25 (43.85%) out of 57 CD-SNPs lies in evolutionarily constrained genetic element regions. In follow-up phases, through computational (CADD, GWAVA, and FATHMM algorithms) deleterious intensity measurements, we have discovered that 42 (3.94%) out of 1065 variants (57 CD-lead and 1008-linked SNPs; r2 ≥ 0.8) are differentially deleterious in nature to CD. Further functional scrutinization of these CD variants by public domain eQTL mapping, gene expression, knockout mouse model, and pathway analyses revealed that deleterious SNPs of CCR2 gene influences its expression levels and may also elicit a cascade of T-cell-mediated immunological events leading to intestinal gluten intolerance in genetically susceptible individuals. This study demonstrates the utility of integrated in silico analysis of annotations, gene expression, and pathways in prioritizing the potential complex disease variants from large-scale open source genomic data. J. Cell. Biochem. 118: 2193-2207, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Babajan Banaganapalli
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.,Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Omran Rashidi
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.,Department of Clinical Biochemistry, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Omar I Saadah
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.,Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Jun Wang
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Imran Ali Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Jumana Y Al-Aama
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.,Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Noor Ahmad Shaik
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Ramu Elango
- Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.,Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
40
|
Hwang IY, Park C, Harrison K, Kehrl JH. Normal Thymocyte Egress, T Cell Trafficking, and CD4 + T Cell Homeostasis Require Interactions between RGS Proteins and Gα i2. THE JOURNAL OF IMMUNOLOGY 2017; 198:2721-2734. [PMID: 28235863 DOI: 10.4049/jimmunol.1601433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/24/2017] [Indexed: 11/19/2022]
Abstract
Adaptive immunity depends on mature thymocytes leaving the thymus to enter the bloodstream and the trafficking of T cells through lymphoid organs. Both of these require heterotrimeric Gαi protein signaling, whose intensity and duration are controlled by the regulator of G protein signaling (RGS) proteins. In this study, we show that RGS protein/Gαi2 interactions are essential for normal thymocyte egress, T cell trafficking, and homeostasis. Mature thymocytes with a Gαi2 mutation that disables RGS protein binding accumulated in the perivascular channels of thymic corticomedullary venules. Severe reductions in peripheral naive CD4+ T cells and regulatory T cells occurred. The mutant CD4+ T cells adhered poorly to high endothelial venules and exhibited defects in lymph node entrance and egress. The kinetics of chemokine receptor signaling were disturbed, including chemokine- induced integrin activation. Despite the thymic and lymph node egress defects, sphingosine-1-phosphate signaling was not obviously perturbed. This study reveals how RGS proteins modulate Gαi2 signaling to facilitate thymocyte egress and T cell trafficking.
Collapse
Affiliation(s)
- Il-Young Hwang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Chung Park
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kathleen Harrison
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - John H Kehrl
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
41
|
Weigt SS, Wang X, Palchevskiy V, Gregson AL, Patel N, DerHovanessian A, Shino MY, Sayah DM, Birjandi S, Lynch JP, Saggar R, Ardehali A, Ross DJ, Palmer SM, Elashoff D, Belperio JA. Gene Expression Profiling of Bronchoalveolar Lavage Cells Preceding a Clinical Diagnosis of Chronic Lung Allograft Dysfunction. PLoS One 2017; 12:e0169894. [PMID: 28103284 PMCID: PMC5245825 DOI: 10.1371/journal.pone.0169894] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 12/22/2016] [Indexed: 11/18/2022] Open
Abstract
Background Chronic Lung Allograft Dysfunction (CLAD) is the main limitation to long-term survival after lung transplantation. Although CLAD is usually not responsive to treatment, earlier identification may improve treatment prospects. Methods In a nested case control study, 1-year post transplant surveillance bronchoalveolar lavage (BAL) fluid samples were obtained from incipient CLAD (n = 9) and CLAD free (n = 8) lung transplant recipients. Incipient CLAD cases were diagnosed with CLAD within 2 years, while controls were free from CLAD for at least 4 years following bronchoscopy. Transcription profiles in the BAL cell pellets were assayed with the HG-U133 Plus 2.0 microarray (Affymetrix). Differential gene expression analysis, based on an absolute fold change (incipient CLAD vs no CLAD) >2.0 and an unadjusted p-value ≤0.05, generated a candidate list containing 55 differentially expressed probe sets (51 up-regulated, 4 down-regulated). Results The cell pellets in incipient CLAD cases were skewed toward immune response pathways, dominated by genes related to recruitment, retention, activation and proliferation of cytotoxic lymphocytes (CD8+ T-cells and natural killer cells). Both hierarchical clustering and a supervised machine learning tool were able to correctly categorize most samples (82.3% and 94.1% respectively) into incipient CLAD and CLAD-free categories. Conclusions These findings suggest that a pathobiology, similar to AR, precedes a clinical diagnosis of CLAD. A larger prospective investigation of the BAL cell pellet transcriptome as a biomarker for CLAD risk stratification is warranted.
Collapse
Affiliation(s)
- S. Samuel Weigt
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- * E-mail:
| | - Xiaoyan Wang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Vyacheslav Palchevskiy
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Aric L. Gregson
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Naman Patel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Ariss DerHovanessian
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Michael Y. Shino
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - David M. Sayah
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Shirin Birjandi
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Joseph P. Lynch
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Rajan Saggar
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Abbas Ardehali
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - David J. Ross
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Scott M. Palmer
- Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - David Elashoff
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - John A. Belperio
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| |
Collapse
|
42
|
Druey KM. Emerging Roles of Regulators of G Protein Signaling (RGS) Proteins in the Immune System. Adv Immunol 2017; 136:315-351. [PMID: 28950950 DOI: 10.1016/bs.ai.2017.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Kirk M Druey
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States.
| |
Collapse
|
43
|
Leigh R, Mostafa MM, King EM, Rider CF, Shah S, Dumonceaux C, Traves SL, McWhae A, Kolisnik T, Kooi C, Slater DM, Kelly MM, Bieda M, Miller-Larsson A, Newton R. An inhaled dose of budesonide induces genes involved in transcription and signaling in the human airways: enhancement of anti- and proinflammatory effector genes. Pharmacol Res Perspect 2016; 4:e00243. [PMID: 28116096 PMCID: PMC5242176 DOI: 10.1002/prp2.243] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/03/2016] [Indexed: 12/27/2022] Open
Abstract
Although inhaled glucocorticoids, or corticosteroids (ICS), are generally effective in asthma, understanding their anti‐inflammatory actions in vivo remains incomplete. To characterize glucocorticoid‐induced modulation of gene expression in the human airways, we performed a randomized placebo‐controlled crossover study in healthy male volunteers. Six hours after placebo or budesonide inhalation, whole blood, bronchial brushings, and endobronchial biopsies were collected. Microarray analysis of biopsy RNA, using stringent (≥2‐fold, 5% false discovery rate) or less stringent (≥1.25‐fold, P ≤ 0.05) criteria, identified 46 and 588 budesonide‐induced genes, respectively. Approximately two third of these genes are transcriptional regulators (KLF9, PER1, TSC22D3, ZBTB16), receptors (CD163, CNR1, CXCR4, LIFR, TLR2), or signaling genes (DUSP1, NFKBIA, RGS1, RGS2, ZFP36). Listed genes were qPCR verified. Expression of anti‐inflammatory and other potentially beneficial genes is therefore confirmed and consistent with gene ontology (GO) terms for negative regulation of transcription and gene expression. However, GO terms for transcription, signaling, metabolism, proliferation, inflammatory responses, and cell movement were also associated with the budesonide‐induced genes. The most enriched functional cluster indicates positive regulation of proliferation, locomotion, movement, and migration. Moreover, comparison with the budesonide‐induced expression profile in primary human airway epithelial cells shows considerable cell type specificity. In conclusion, increased expression of multiple genes, including the transcriptional repressor, ZBTB16, that reduce inflammatory signaling and gene expression, occurs in the airways and blood and may contribute to the therapeutic efficacy of ICS. This provides a previously lacking insight into the in vivo effects of ICS and should promote strategies to improve glucocorticoid efficacy in inflammatory diseases.
Collapse
Affiliation(s)
- Richard Leigh
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Mahmoud M Mostafa
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Elizabeth M King
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Christopher F Rider
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Suharsh Shah
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Curtis Dumonceaux
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Suzanne L Traves
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Andrew McWhae
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Tyler Kolisnik
- Alberta Children's Hospital Research Institute University of Calgary Calgary Alberta T2N 4Z6
| | - Cora Kooi
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Donna M Slater
- Alberta Children's Hospital Research Institute University of Calgary Calgary Alberta T2N 4Z6
| | - Margaret M Kelly
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| | - Mark Bieda
- Alberta Children's Hospital Research Institute University of Calgary Calgary Alberta T2N 4Z6
| | | | - Robert Newton
- Airways Inflammation Research Group Snyder Institute for Chronic Diseases University of Calgary Calgary Alberta T2N 4Z6
| |
Collapse
|
44
|
Kehrl JH. The impact of RGS and other G-protein regulatory proteins on Gαi-mediated signaling in immunity. Biochem Pharmacol 2016; 114:40-52. [PMID: 27071343 DOI: 10.1016/j.bcp.2016.04.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/08/2016] [Indexed: 01/30/2023]
Abstract
Leukocyte chemoattractant receptors are members of the G-protein coupled receptor (GPCR) family. Signaling downstream of these receptors directs the localization, positioning and homeostatic trafficking of leukocytes; as well as their recruitment to, and their retention at, inflammatory sites. Ligand induced changes in the molecular conformation of chemoattractant receptors results in the engagement of heterotrimeric G-proteins, which promotes α subunits to undergo GTP/GDP exchange. This results in the functional release of βγ subunits from the heterotrimers, thereby activating downstream effector molecules, which initiate leukocyte polarization, gradient sensing, and directional migration. Pertussis toxin ADP ribosylates Gαi subunits and prevents chemoattractant receptors from triggering Gαi nucleotide exchange. The use of pertussis toxin revealed the essential importance of Gαi subunit nucleotide exchange for chemoattractant receptor signaling. More recent studies have identified a range of regulatory mechanisms that target these receptors and their associated heterotrimeric G-proteins, thereby helping to control the magnitude, kinetics, and duration of signaling. A failure in these regulatory pathways can lead to impaired receptor signaling and immunopathology. The analysis of mice with targeted deletions of Gαi isoforms as well as some of these G-protein regulatory proteins is providing insights into their roles in chemoattractant receptor signaling.
Collapse
Affiliation(s)
- John H Kehrl
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 2089, United States.
| |
Collapse
|
45
|
Salaga M, Storr M, Martemyanov KA, Fichna J. RGS proteins as targets in the treatment of intestinal inflammation and visceral pain: New insights and future perspectives. Bioessays 2016; 38:344-54. [PMID: 26817719 PMCID: PMC4916644 DOI: 10.1002/bies.201500118] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Regulators of G protein signaling (RGS) proteins provide timely termination of G protein-coupled receptor (GPCR) responses. Serving as a central control point in GPCR signaling cascades, RGS proteins are promising targets for drug development. In this review, we discuss the involvement of RGS proteins in the pathophysiology of the gastrointestinal inflammation and their potential to become a target for anti-inflammatory drugs. Specifically, we evaluate the emerging evidence for modulation of selected receptor families: opioid, cannabinoid and serotonin by RGS proteins. We discuss how the regulation of RGS protein level and activity may modulate immunological pathways involved in the development of intestinal inflammation. Finally, we propose that RGS proteins may serve as a prognostic factor for survival rate in colorectal cancer. The ideas introduced in this review set a novel conceptual framework for the utilization of RGS proteins in the treatment of gastrointestinal inflammation, a growing major concern worldwide.
Collapse
Affiliation(s)
- Maciej Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Poland
| | - Martin Storr
- Walter Brendel Center of Experimental Medicine, University of Munich, Germany
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
- Corresponding authors: J.F. Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland, Phone: ++48 42 272 57 07, Fax: ++48 42 272 56 94, . K.A.M., Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way C347, Jupiter, FL 33458, USA, Phone: ++1 561 228 2770,
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Poland
- Corresponding authors: J.F. Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland, Phone: ++48 42 272 57 07, Fax: ++48 42 272 56 94, . K.A.M., Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way C347, Jupiter, FL 33458, USA, Phone: ++1 561 228 2770,
| |
Collapse
|
46
|
The autoimmunity-associated gene RGS1 affects the frequency of T follicular helper cells. Genes Immun 2016; 17:228-38. [PMID: 27029527 PMCID: PMC4892947 DOI: 10.1038/gene.2016.16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/19/2016] [Accepted: 02/25/2016] [Indexed: 12/14/2022]
Abstract
RGS1 (regulator of G-protein signaling 1) has been associated with multiple autoimmune disorders including type I diabetes. RGS1 desensitizes the chemokine receptors CCR7 and CXCR4 that are critical to the localization of T and B cells in lymphoid organs. To explore how RGS1 variation contributes to autoimmunity, we generated Rgs1 knockdown (KD) mice in the nonobese diabetic (NOD) model for type I diabetes. We found that Rgs1 KD increased the size of germinal centers, but decreased the frequency of T follicular helper (TFH) cells. We show that loss of Rgs1 in T cells had both a T cell-intrinsic effect on migration and TFH cell frequency, and an indirect effect on B-cell migration and germinal center formation. Notably, several recent publications described an increase in circulating TFH cells in patients with type I diabetes, suggesting this cell population is involved in pathogenesis. Though Rgs1 KD was insufficient to alter diabetes frequency in the NOD model, our findings raise the possibility that RGS1 plays a role in autoimmunity owing to its function in TFH cells. This mechanistic link, although speculative at this time, would lend support to the notion that TFH cells are key participants in autoimmunity and could explain the association of RGS1 with several immune-mediated diseases.
Collapse
|
47
|
Meta-Analysis on Associations of RGS1 and IL12A Polymorphisms with Celiac Disease Risk. Int J Mol Sci 2016; 17:457. [PMID: 27043536 PMCID: PMC4848913 DOI: 10.3390/ijms17040457] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/17/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of celiac disease (CD) has been related to polymorphisms in the regulator of G-protein signaling 1 (RGS1) and interleukin-12 A (IL12A) genes, but the existing findings are inconsistent. Our aim is to investigate the associations of two single-nucleotide polymorphisms (SNPs) (rs2816316 in RGS1 and rs17810546 in IL12A) with CD risk using meta-analysis. We searched PubMed and Web of Science on RGS1 rs2816316 and IL12A rs17810546 with CD risk. Odds ratio (OR) and 95% confidence interval (CI) of each SNP were estimated. All statistical analyses were performed on Stata 12.0. A total of seven studies were retrieved and analyzed. The available data indicated the minor allele C of rs2816316 was negatively associated with CD (C vs. A: OR = 0.77, 95% CI = 0.74-0.80), and a positive association was found for the minor allele G of rs17810546 (G vs. A: OR = 1.37, 95% CI = 1.31-1.43). The co-dominant model of genotype effect confirmed the significant associations between RGS1 rs2816316/IL12A rs17810546 and CD. No evidence of publication bias was observed. Our meta-analysis supports the associations of RGS1 and IL12A with CD and strongly calls for further studies to better understand the roles of RGS1 and IL12A in the pathogenesis of CD.
Collapse
|
48
|
Lee JK, Bou Dagher J. Regulator of G-protein Signaling (RGS)1 and RGS10 Proteins as Potential Drug Targets for Neuroinflammatory and Neurodegenerative Diseases. AAPS JOURNAL 2016; 18:545-9. [PMID: 26902301 DOI: 10.1208/s12248-016-9883-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/02/2016] [Indexed: 01/25/2023]
Abstract
Regulator of G-protein signaling (RGS) proteins were originally identified as negative regulators of G-protein-coupled receptor (GPCR) signaling via their GTPase-accelerating protein (GAP) activity. All RGS proteins contain evolutionarily conserved RGS domain; however, they differ in their size and regulatory domains. RGS1 and RGS10 are smaller than other RGS proteins, and their functions involve various inflammatory responses including autoimmune responses in both the periphery and the central nervous system (CNS). Neuroinflammation is the chronic inflammatory response in the CNS. Acute inflammatory response in the CNS is believed to be beneficial by involving the neuroprotective actions of immune cells in the brain, particularly microglia, to limit tissue damage and to aid in neuronal repair. However, chronically elevated levels of cytokines serve to maintain activation of abundant numbers of immune cells potentiating prolonged inflammatory responses and creating an environment of oxidative stress, which further hastens oxidative damage of neurons. In this review, we describe the implications and features of RGS proteins (specifically RGS1 and RGS10) in neuroinflammation and neurodegenerative diseases. We will discuss the experimental and epidemiological evidence on the benefits of anti-inflammatory interventions by targeting RGS1 and/or RGS10 protein function or expression in order to delay or attenuate the progression of neurodegeneration, particularly in multiple sclerosis (MS) and Parkinson's disease (PD).
Collapse
Affiliation(s)
- Jae-Kyung Lee
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Dr., Athens, Georgia, 30602, USA.
| | - Josephine Bou Dagher
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Dr., Athens, Georgia, 30602, USA
| |
Collapse
|
49
|
Xie Z, Chan EC, Druey KM. R4 Regulator of G Protein Signaling (RGS) Proteins in Inflammation and Immunity. AAPS JOURNAL 2015; 18:294-304. [PMID: 26597290 DOI: 10.1208/s12248-015-9847-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/11/2015] [Indexed: 11/30/2022]
Abstract
G protein-coupled receptors (GPCRs) have important functions in both innate and adaptive immunity, with the capacity to bridge interactions between the two arms of the host responses to pathogens through direct recognition of secreted microbial products or the by-products of host cells damaged by pathogen exposure. In the mid-1990s, a large group of intracellular proteins was discovered, the regulator of G protein signaling (RGS) family, whose main, but not exclusive, function appears to be to constrain the intensity and duration of GPCR signaling. The R4/B subfamily--the focus of this review--includes RGS1-5, 8, 13, 16, 18, and 21, which are the smallest RGS proteins in size, with the exception of RGS3. Prominent roles in the trafficking of B and T lymphocytes and macrophages have been described for RGS1, RGS13, and RGS16, while RGS18 appears to control platelet and osteoclast functions. Additional G protein independent functions of RGS13 have been uncovered in gene expression in B lymphocytes and mast cell-mediated allergic reactions. In this review, we discuss potential physiological roles of this RGS protein subfamily, primarily in leukocytes having central roles in immune and inflammatory responses. We also discuss approaches to target RGS proteins therapeutically, which represents a virtually untapped strategy to combat exaggerated immune responses leading to inflammation.
Collapse
Affiliation(s)
- Zhihui Xie
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA
| | - Eunice C Chan
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA
| | - Kirk M Druey
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
50
|
Giordano M, Henin C, Maurizio J, Imbratta C, Bourdely P, Buferne M, Baitsch L, Vanhille L, Sieweke MH, Speiser DE, Auphan-Anezin N, Schmitt-Verhulst AM, Verdeil G. Molecular profiling of CD8 T cells in autochthonous melanoma identifies Maf as driver of exhaustion. EMBO J 2015; 34:2042-58. [PMID: 26139534 DOI: 10.15252/embj.201490786] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/08/2015] [Indexed: 01/12/2023] Open
Abstract
T cells infiltrating neoplasms express surface molecules typical of chronically virus-stimulated T cells, often termed "exhausted" T cells. We compared the transcriptome of "exhausted" CD8 T cells infiltrating autochthonous melanomas to those of naïve and acutely stimulated CD8 T cells. Despite strong similarities between transcriptional signatures of tumor- and virus-induced exhausted CD8 T cells, notable differences appeared. Among transcriptional regulators, Nr4a2 and Maf were highly overexpressed in tumor-exhausted T cells and significantly upregulated in CD8 T cells from human melanoma metastases. Transduction of murine tumor-specific CD8 T cells to express Maf partially reproduced the transcriptional program associated with tumor-induced exhaustion. Upon adoptive transfer, the transduced cells showed normal homeostasis but failed to accumulate in tumor-bearing hosts and developed defective anti-tumor effector responses. We further identified TGFβ and IL-6 as main inducers of Maf expression in CD8 T cells and showed that Maf-deleted tumor-specific CD8 T cells were much more potent to restrain tumor growth in vivo. Therefore, the melanoma microenvironment contributes to skewing of CD8 T cell differentiation programs, in part by TGFβ/IL-6-mediated induction of Maf.
Collapse
Affiliation(s)
- Marilyn Giordano
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Coralie Henin
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Julien Maurizio
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Claire Imbratta
- Clinical Tumor Biology & Immunotherapy Group, Department of Oncology and Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
| | - Pierre Bourdely
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Michel Buferne
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Lukas Baitsch
- Clinical Tumor Biology & Immunotherapy Group, Department of Oncology and Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
| | - Laurent Vanhille
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Michael H Sieweke
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
| | - Daniel E Speiser
- Clinical Tumor Biology & Immunotherapy Group, Department of Oncology and Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
| | - Nathalie Auphan-Anezin
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Anne-Marie Schmitt-Verhulst
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Grégory Verdeil
- Centre d'Immunologie de Marseille-Luminy (CIML), UM2 Aix-Marseille Université, Marseille Cedex 9, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| |
Collapse
|