1
|
Boschiero C, Beshah E, Zhu X, Tuo W, Liu GE. Profiling Genome-Wide Methylation Patterns in Cattle Infected with Ostertagia ostertagi. Int J Mol Sci 2024; 26:89. [PMID: 39795948 PMCID: PMC11719486 DOI: 10.3390/ijms26010089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/13/2025] Open
Abstract
DNA methylation (DNAm) regulates gene expression and genomic imprinting. This study aimed to investigate the effect of gastrointestinal (GI) nematode infection on host DNAm. Helminth-free Holstein steers were either infected with Ostertagia ostertagi (the brown stomach worm) or given tap water only as a control. Animals were euthanized 30 days post-infection, and tissues were collected at necropsy. We conducted epigenome-wide profiling using a mammalian methylation array to explore the impact of infection on methylation patterns in the mucosa from abomasal fundus (FUN), pylorus (PYL), draining lymph nodes (dLNs), and the duodenum (DUO). The analysis covered 31,107 cattle CpGs of 5082 genes and revealed infection-driven, tissue-specific, differential methylation patterns. A total of 389 shared and 2770 tissue-specific, differentially methylated positions (DMPs) were identified in dLN and FUN, particularly in genes associated with immune responses. The shared DMPs were found in 263 genes, many of which are involved in immune responses. Furthermore, 282, 244, 52, and 24 differentially methylated regions (DMRs) were observed in dLN, FUN, PYL, and DUO, respectively. More hypomethylated DMRs were detected in dLN and FUN, while more hypermethylated DMRs were found in PYL and DUO. Genes carrying DMPs and DMRs and enriched pathways relating to immune functions/responses were detected in infected animals, indicating a link between DNA methylation and the infection. The data may implicate a crucial role of DNAm in regulating the nature/strength of host immunity to infection and contribute to a deeper understanding of the epigenetic regulatory landscape in cattle infected by GI nematodes.
Collapse
Affiliation(s)
- Clarissa Boschiero
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Ethiopia Beshah
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Xiaoping Zhu
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Wenbin Tuo
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - George E. Liu
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| |
Collapse
|
2
|
Li K, Desai R, Scott RT, Steele JR, Machado M, Demharter S, Hoarfrost A, Braun JL, Fajardo VA, Sanders LM, Costes SV. Explainable machine learning identifies multi-omics signatures of muscle response to spaceflight in mice. NPJ Microgravity 2023; 9:90. [PMID: 38092777 PMCID: PMC10719374 DOI: 10.1038/s41526-023-00337-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
The adverse effects of microgravity exposure on mammalian physiology during spaceflight necessitate a deep understanding of the underlying mechanisms to develop effective countermeasures. One such concern is muscle atrophy, which is partly attributed to the dysregulation of calcium levels due to abnormalities in SERCA pump functioning. To identify potential biomarkers for this condition, multi-omics data and physiological data available on the NASA Open Science Data Repository (osdr.nasa.gov) were used, and machine learning methods were employed. Specifically, we used multi-omics (transcriptomic, proteomic, and DNA methylation) data and calcium reuptake data collected from C57BL/6 J mouse soleus and tibialis anterior tissues during several 30+ day-long missions on the international space station. The QLattice symbolic regression algorithm was introduced to generate highly explainable models that predict either experimental conditions or calcium reuptake levels based on multi-omics features. The list of candidate models established by QLattice was used to identify key features contributing to the predictive capability of these models, with Acyp1 and Rps7 proteins found to be the most predictive biomarkers related to the resilience of the tibialis anterior muscle in space. These findings could serve as targets for future interventions aiming to reduce the extent of muscle atrophy during space travel.
Collapse
Affiliation(s)
- Kevin Li
- KBR, Moffett Field, CA, USA
- NASA Space Life Sciences Training Program, Moffett Field, CA, USA
| | - Riya Desai
- College of Letters and Science, University of California at Davis, Davis, CA, USA
| | - Ryan T Scott
- KBR, Moffett Field, CA, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Joel Ricky Steele
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Blue Marble Space, Seattle, WA, USA
| | | | | | | | - Jessica L Braun
- Department of Kinesiology, Centre for Bone and Muscle Health, Brock University, St. Catharines, Canada
| | - Val A Fajardo
- Department of Kinesiology, Centre for Bone and Muscle Health, Brock University, St. Catharines, Canada
| | - Lauren M Sanders
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
- Blue Marble Space, Seattle, WA, USA.
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
| |
Collapse
|
3
|
Galazo MJ, Sweetser DA, Macklis JD. Tle4 controls both developmental acquisition and early post-natal maturation of corticothalamic projection neuron identity. Cell Rep 2023; 42:112957. [PMID: 37561632 PMCID: PMC10542749 DOI: 10.1016/j.celrep.2023.112957] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 04/21/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Identities of distinct neuron subtypes are specified during embryonic development, then maintained during post-natal maturation. In cerebral cortex, mechanisms controlling early acquisition of neuron-subtype identities have become increasingly understood. However, mechanisms controlling neuron-subtype identity stability during post-natal maturation are largely unexplored. We identify that Tle4 is required for both early acquisition and post-natal stability of corticothalamic neuron-subtype identity. Embryonically, Tle4 promotes acquisition of corticothalamic identity and blocks emergence of core characteristics of subcerebral/corticospinal projection neuron identity, including gene expression and connectivity. During the first post-natal week, when corticothalamic innervation is ongoing, Tle4 is required to stabilize corticothalamic neuron identity, limiting interference from differentiation programs of developmentally related neuron classes. We identify a deacetylation-based epigenetic mechanism by which TLE4 controls Fezf2 expression level by corticothalamic neurons. This contributes to distinction of cortical output subtypes and ensures identity stability for appropriate maturation of corticothalamic neurons.
Collapse
Affiliation(s)
- Maria J Galazo
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - David A Sweetser
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
4
|
Yu G, Chen Y, Hu Y, Zhou Y, Ding X, Zhou X. Roles of transducin-like enhancer of split (TLE) family proteins in tumorigenesis and immune regulation. Front Cell Dev Biol 2022; 10:1010639. [PMID: 36438567 PMCID: PMC9692235 DOI: 10.3389/fcell.2022.1010639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/31/2022] [Indexed: 08/16/2023] Open
Abstract
Mammalian transducin-like enhancer of split family proteins (TLEs) are homologous to Drosophila Groucho (Gro) and are essential transcriptional repressors. Seven TLE family members, TLE1-7, have been identified to date. These proteins do not bind DNA directly; instead, they bind a set of transcription factors and thereby inhibit target gene expression. Loss of TLEs in mice usually leads to defective early development; however, TLE functions in developmentally mature cells are unclear. Recent studies have revealed that TLEs are dysregulated in certain human cancer types and may function as oncogenes or tumor suppressors in different contexts. TLE levels also affect the efficacy of cancer treatments and the development of drug resistance. In addition, TLEs play critical roles in the development and function of immune cells, including macrophages and lymphocytes. In this review, we provide updates on the expression, function, and mechanism of TLEs; discuss the roles played by TLEs in tumorigenesis and the inflammatory response; and elaborate on several TLE-associated signaling pathways, including the Notch, Wnt, and MAPK pathways. Finally, we discuss potential strategies for targeting TLEs in cancer therapy.
Collapse
Affiliation(s)
- Guiping Yu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, China
| | - Yiqi Chen
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Yuwen Hu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Yan Zhou
- Department of Periodontology, The Affiliated Nantong Stomatological Hospital of Nantong University, Nantong, China
| | - Xiaoling Ding
- Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaorong Zhou
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| |
Collapse
|
5
|
Murty T, Mackall CL. Gene editing to enhance the efficacy of cancer cell therapies. Mol Ther 2021; 29:3153-3162. [PMID: 34673274 PMCID: PMC8571170 DOI: 10.1016/j.ymthe.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/18/2022] Open
Abstract
Adoptive T cell therapies have shown impressive signals of activity, but their clinical impact could be enhanced by technologies to increase T cell potency and diminish the cost and labor involved in manufacturing these products. Gene editing platforms are under study in this arena to (1) enhance immune cell potency by knocking out molecules that inhibit immune responses; (2) deliver genetic payloads into precise genomic locations and thereby enhance safety and/or improve the gene expression profile by leveraging physiologic promoters, enhancers, and repressors; and (3) enable off-the-shelf therapies by preventing alloreactivity and immune rejection. This review discusses gene editing approaches that have been the best studied in the context of human T cells and adoptive T cell therapies, summarizing their current status and near-term potential for translation.
Collapse
Affiliation(s)
- Tara Murty
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA; Program in Biophysics, Stanford University, Stanford, CA, USA; Medical Scientist Training Program, Stanford University, Stanford, CA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Golub R. The Notch signaling pathway involvement in innate lymphoid cell biology. Biomed J 2020; 44:133-143. [PMID: 33863682 PMCID: PMC8178581 DOI: 10.1016/j.bj.2020.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/30/2022] Open
Abstract
The role of Notch in the immune system was first described in the late 90s. Reports revealed that Notch is one of the most conserved developmental pathways involved in diverse biological processes such as the development, differentiation, survival and functions of many immune populations. Here, we provide an extended view of the pleiotropic effects of the Notch signaling on the innate lymphoid cell (ILC) biology. We review the current knowledge on Notch signaling in the regulation of ILC differentiation, plasticity and functions in diverse tissue types and at both the fetal and adult developmental stages. ILCs are early responder cells that secrete a large panel of cytokines after stimulation. By controlling the abundance of ILCs and the specificity of their release, the Notch pathway is also implicated in the regulation of their functions. The Notch pathway is therefore an important player in both ILC cell fate decision and ILC immune response.
Collapse
Affiliation(s)
- Rachel Golub
- Unit of Lymphocytes and Immunity, Department of Immunology, Institut Pasteur, Paris, France.
| |
Collapse
|
7
|
Wang D, Prager BC, Gimple RC, Aguilar B, Alizadeh D, Tang H, Lv D, Starr R, Brito A, Wu Q, Kim LJY, Qiu Z, Lin P, Lorenzini MH, Badie B, Forman SJ, Xie Q, Brown CE, Rich JN. CRISPR Screening of CAR T Cells and Cancer Stem Cells Reveals Critical Dependencies for Cell-Based Therapies. Cancer Discov 2020; 11:1192-1211. [PMID: 33328215 DOI: 10.1158/2159-8290.cd-20-1243] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/02/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) contains self-renewing GBM stem cells (GSC) potentially amenable to immunologic targeting, but chimeric antigen receptor (CAR) T-cell therapy has demonstrated limited clinical responses in GBM. Here, we interrogated molecular determinants of CAR-mediated GBM killing through whole-genome CRISPR screens in both CAR T cells and patient-derived GSCs. Screening of CAR T cells identified dependencies for effector functions, including TLE4 and IKZF2. Targeted knockout of these genes enhanced CAR antitumor efficacy. Bulk and single-cell RNA sequencing of edited CAR T cells revealed transcriptional profiles of superior effector function and inhibited exhaustion responses. Reciprocal screening of GSCs identified genes essential for susceptibility to CAR-mediated killing, including RELA and NPLOC4, the knockout of which altered tumor-immune signaling and increased responsiveness of CAR therapy. Overall, CRISPR screening of CAR T cells and GSCs discovered avenues for enhancing CAR therapeutic efficacy against GBM, with the potential to be extended to other solid tumors. SIGNIFICANCE: Reciprocal CRISPR screening identified genes in both CAR T cells and tumor cells regulating the potency of CAR T-cell cytotoxicity, informing molecular targeting strategies to potentiate CAR T-cell antitumor efficacy and elucidate genetic modifications of tumor cells in combination with CAR T cells to advance immuno-oncotherapy.This article is highlighted in the In This Issue feature, p. 995.
Collapse
Affiliation(s)
- Dongrui Wang
- T Cell Therapeutics Research Labs, Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Briana C Prager
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Cleveland Clinic Lerner College of Medicine at Cleveland Clinic and Case Western Reserve University, Cleveland, Ohio.,Sanford Consortium for Regenerative Medicine, La Jolla, California
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Sanford Consortium for Regenerative Medicine, La Jolla, California.,Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Brenda Aguilar
- T Cell Therapeutics Research Labs, Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Darya Alizadeh
- T Cell Therapeutics Research Labs, Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Hongzhen Tang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
| | - Deguan Lv
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Sanford Consortium for Regenerative Medicine, La Jolla, California
| | - Renate Starr
- T Cell Therapeutics Research Labs, Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Alfonso Brito
- T Cell Therapeutics Research Labs, Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Qiulian Wu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Sanford Consortium for Regenerative Medicine, La Jolla, California
| | - Leo J Y Kim
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Sanford Consortium for Regenerative Medicine, La Jolla, California.,Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Zhixin Qiu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Sanford Consortium for Regenerative Medicine, La Jolla, California
| | - Peng Lin
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
| | - Michael H Lorenzini
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Sanford Consortium for Regenerative Medicine, La Jolla, California
| | - Behnam Badie
- Division of Neurosurgery, Department of Surgery, City of Hope, Duarte, California
| | - Stephen J Forman
- T Cell Therapeutics Research Labs, Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Qi Xie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China. .,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
| | - Christine E Brown
- T Cell Therapeutics Research Labs, Cellular Immunotherapy Center, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California.
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California. .,Sanford Consortium for Regenerative Medicine, La Jolla, California.,University of Pittsburgh Medical Center Hillman Cancer Center, Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
8
|
Eyring KR, Pedersen BS, Maclean KN, Stabler SP, Yang IV, Schwartz DA. Methylene-tetrahydrofolate reductase contributes to allergic airway disease. PLoS One 2018; 13:e0190916. [PMID: 29329322 PMCID: PMC5766142 DOI: 10.1371/journal.pone.0190916] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 12/22/2017] [Indexed: 12/01/2022] Open
Abstract
Rationale Environmental exposures strongly influence the development and progression of asthma. We have previously demonstrated that mice exposed to a diet enriched with methyl donors during vulnerable periods of fetal development can enhance the heritable risk of allergic airway disease through epigenetic changes. There is conflicting evidence on the role of folate (one of the primary methyl donors) in modifying allergic airway disease. Objectives We hypothesized that blocking folate metabolism through the loss of methylene-tetrahydrofolate reductase (Mthfr) activity would reduce the allergic airway disease phenotype through epigenetic mechanisms. Methods Allergic airway disease was induced in C57BL/6 and C57BL/6Mthfr-/- mice through house dust mite (HDM) exposure. Airway inflammation and airway hyperresponsiveness (AHR) were measured between the two groups. Gene expression and methylation profiles were generated for whole lung tissue. Disease and molecular outcomes were evaluated in C57BL/6 and C57BL/6Mthfr-/- mice supplemented with betaine. Measurements and main results Loss of Mthfr alters single carbon metabolite levels in the lung and serum including elevated homocysteine and cystathionine and reduced methionine. HDM-treated C57BL/6Mthfr-/- mice demonstrated significantly less airway hyperreactivity (AHR) compared to HDM-treated C57BL/6 mice. Furthermore, HDM-treated C57BL/6Mthfr-/- mice compared to HDM-treated C57BL/6 mice have reduced whole lung lavage (WLL) cellularity, eosinophilia, and Il-4/Il-5 cytokine concentrations. Betaine supplementation reversed parts of the HDM-induced allergic airway disease that are modified by Mthfr loss. 737 genes are differentially expressed and 146 regions are differentially methylated in lung tissue from HDM-treated C57BL/6Mthfr-/- mice and HDM-treated C57BL/6 mice. Additionally, analysis of methylation/expression relationships identified 503 significant correlations. Conclusion Collectively, these findings indicate that the loss of folate as a methyl donor is a modifier of allergic airway disease, and that epigenetic and expression changes correlate with this modification. Further investigation into the mechanisms that drive this observation is warranted.
Collapse
Affiliation(s)
- Kenneth R. Eyring
- Department of Medicine, School of Medicine, University of Colorado, Aurora, CO, United States of America
| | - Brent S. Pedersen
- Department of Medicine, School of Medicine, University of Colorado, Aurora, CO, United States of America
| | - Kenneth N. Maclean
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO, United States of America
| | - Sally P. Stabler
- Department of Medicine, School of Medicine, University of Colorado, Aurora, CO, United States of America
| | - Ivana V. Yang
- Department of Medicine, School of Medicine, University of Colorado, Aurora, CO, United States of America
| | - David A. Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, CO, United States of America
- Department of Immunology, School of Medicine, University of Colorado, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
9
|
Xu Y, Zhao F, Qiu Q, Chen K, Wei J, Kong Q, Gao B, Melo-Cardenas J, Zhang B, Zhang J, Song J, Zhang DD, Zhang J, Fan Y, Li H, Fang D. The ER membrane-anchored ubiquitin ligase Hrd1 is a positive regulator of T-cell immunity. Nat Commun 2016; 7:12073. [PMID: 27417417 PMCID: PMC4947160 DOI: 10.1038/ncomms12073] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/25/2016] [Indexed: 01/16/2023] Open
Abstract
Identification of positive regulators of T-cell immunity induced during autoimmune diseases is critical for developing novel therapies. The endoplasmic reticulum resident ubiquitin ligase Hrd1 has recently emerged as a critical regulator of dendritic cell antigen presentation, but its role in T-cell immunity is unknown. Here we show that genetic deletion of Hrd1 in mice inhibits T-cell proliferation, production of IL-2, and differentiation of Th1 and Th17 cells, and consequently protects mice from experimental autoimmune encephalomyelitis. Hrd1 facilitates T-cell proliferation by the destruction of cyclin-dependent kinase inhibitor p27(kip1), and deletion of p27(kip1) in Hrd1-null T-cells rescues proliferative capacity but not the production of cytokines, including IL-2, IFN-γ and IL-17. T-cell expression of Hrd1 is higher in patients with multiple sclerosis than in healthy individuals, and knockdown of Hrd1 in human CD4(+) T cells inhibits activation and differentiation to Th1 and Th17 cells. Our study identifies Hrd1 as a previously unappreciated positive regulator of T cells and implies that Hrd1 is a potential therapeutic target for autoimmune diseases.
Collapse
Affiliation(s)
- Yuanming Xu
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Fang Zhao
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Quan Qiu
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
- Allergy Center, Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Kun Chen
- Allergy Center, Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Qingfei Kong
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Beixue Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Johanna Melo-Cardenas
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Bin Zhang
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine-Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Jianxun Song
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizon 85721, USA
| | - Jianing Zhang
- Department of Biochemistry, School of Life Science and Medicine, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Yunping Fan
- Guangdong Provincial Engineering Research Center for Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Huabin Li
- Allergy Center, Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye and ENT Hospital, Fudan University, Shanghai 200031, China
- Guangdong Provincial Engineering Research Center for Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| |
Collapse
|
10
|
The heterogeneity of human CD127(+) innate lymphoid cells revealed by single-cell RNA sequencing. Nat Immunol 2016; 17:451-60. [PMID: 26878113 DOI: 10.1038/ni.3368] [Citation(s) in RCA: 384] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 12/07/2015] [Indexed: 12/13/2022]
Abstract
Innate lymphoid cells (ILCs) are increasingly appreciated as important participants in homeostasis and inflammation. Substantial plasticity and heterogeneity among ILC populations have been reported. Here we have delineated the heterogeneity of human ILCs through single-cell RNA sequencing of several hundreds of individual tonsil CD127(+) ILCs and natural killer (NK) cells. Unbiased transcriptional clustering revealed four distinct populations, corresponding to ILC1 cells, ILC2 cells, ILC3 cells and NK cells, with their respective transcriptomes recapitulating known as well as unknown transcriptional profiles. The single-cell resolution additionally divulged three transcriptionally and functionally diverse subpopulations of ILC3 cells. Our systematic comparison of single-cell transcriptional variation within and between ILC populations provides new insight into ILC biology during homeostasis, with additional implications for dysregulation of the immune system.
Collapse
|