151
|
Puech PH, Bongrand P. Mechanotransduction as a major driver of cell behaviour: mechanisms, and relevance to cell organization and future research. Open Biol 2021; 11:210256. [PMID: 34753321 PMCID: PMC8586914 DOI: 10.1098/rsob.210256] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023] Open
Abstract
How do cells process environmental cues to make decisions? This simple question is still generating much experimental and theoretical work, at the border of physics, chemistry and biology, with strong implications in medicine. The purpose of mechanobiology is to understand how biochemical and physical cues are turned into signals through mechanotransduction. Here, we review recent evidence showing that (i) mechanotransduction plays a major role in triggering signalling cascades following cell-neighbourhood interaction; (ii) the cell capacity to continually generate forces, and biomolecule properties to undergo conformational changes in response to piconewton forces, provide a molecular basis for understanding mechanotransduction; and (iii) mechanotransduction shapes the guidance cues retrieved by living cells and the information flow they generate. This includes the temporal and spatial properties of intracellular signalling cascades. In conclusion, it is suggested that the described concepts may provide guidelines to define experimentally accessible parameters to describe cell structure and dynamics, as a prerequisite to take advantage of recent progress in high-throughput data gathering, computer simulation and artificial intelligence, in order to build a workable, hopefully predictive, account of cell signalling networks.
Collapse
Affiliation(s)
- Pierre-Henri Puech
- Lab Adhesion and Inflammation (LAI), Inserm UMR 1067, CNRS UMR 7333, Aix-Marseille Université UM61, Marseille, France
| | - Pierre Bongrand
- Lab Adhesion and Inflammation (LAI), Inserm UMR 1067, CNRS UMR 7333, Aix-Marseille Université UM61, Marseille, France
| |
Collapse
|
152
|
Vanderwall D, Suresh P, Fu Y, Cho JH, Shaw TI, Mishra A, High AA, Peng J, Li Y. JUMPn: A Streamlined Application for Protein Co-Expression Clustering and Network Analysis in Proteomics. J Vis Exp 2021:10.3791/62796. [PMID: 34747401 PMCID: PMC9185798 DOI: 10.3791/62796] [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] [Indexed: 01/10/2023] Open
Abstract
With recent advances in mass spectrometry-based proteomics technologies, deep profiling of hundreds of proteomes has become increasingly feasible. However, deriving biological insights from such valuable datasets is challenging. Here we introduce a systems biology-based software JUMPn, and its associated protocol to organize the proteome into protein co-expression clusters across samples and protein-protein interaction (PPI) networks connected by modules (e.g., protein complexes). Using the R/Shiny platform, the JUMPn software streamlines the analysis of co-expression clustering, pathway enrichment, and PPI module detection, with integrated data visualization and a user-friendly interface. The main steps of the protocol include installation of the JUMPn software, the definition of differentially expressed proteins or the (dys)regulated proteome, determination of meaningful co-expression clusters and PPI modules, and result visualization. While the protocol is demonstrated using an isobaric labeling-based proteome profile, JUMPn is generally applicable to a wide range of quantitative datasets (e.g., label-free proteomics). The JUMPn software and protocol thus provide a powerful tool to facilitate biological interpretation in quantitative proteomics.
Collapse
Affiliation(s)
- David Vanderwall
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital
| | - Poudel Suresh
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital; Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital
| | - Yingxue Fu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital
| | - Ji-Hoon Cho
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital
| | - Timothy I Shaw
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital; Department of Computational Biology, St. Jude Children's Research Hospital
| | - Ashutosh Mishra
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital
| | - Anthony A High
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital; Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital;
| | - Yuxin Li
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital; Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital;
| |
Collapse
|
153
|
Lewis DA, Ly T. Cell Cycle Entry Control in Naïve and Memory CD8 + T Cells. Front Cell Dev Biol 2021; 9:727441. [PMID: 34692683 PMCID: PMC8526999 DOI: 10.3389/fcell.2021.727441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/07/2021] [Indexed: 12/20/2022] Open
Abstract
CD8+ T cells play important roles in immunity and immuno-oncology. Upon antigen recognition and co-stimulation, naïve CD8+ T cells escape from dormancy to engage in a complex programme of cellular growth, cell cycle entry and differentiation, resulting in rapid proliferation cycles that has the net effect of producing clonally expanded, antigen-specific cytotoxic T lymphocytes (CTLs). A fraction of activated T cells will re-enter dormancy by differentiating into memory T cells, which have essential roles in adaptive immunity. In this review, we discuss the current understanding of cell cycle entry control in CD8+ T cells and crosstalk between these mechanisms and pathways regulating immunological phenotypes.
Collapse
Affiliation(s)
- David A. Lewis
- Ashworth Laboratories, Institute of Immunology and Infectious Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Tony Ly
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
154
|
Mimitou EP, Lareau CA, Chen KY, Zorzetto-Fernandes AL, Hao Y, Takeshima Y, Luo W, Huang TS, Yeung BZ, Papalexi E, Thakore PI, Kibayashi T, Wing JB, Hata M, Satija R, Nazor KL, Sakaguchi S, Ludwig LS, Sankaran VG, Regev A, Smibert P. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat Biotechnol 2021; 39:1246-1258. [PMID: 34083792 PMCID: PMC8763625 DOI: 10.1038/s41587-021-00927-2] [Citation(s) in RCA: 276] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 04/16/2021] [Indexed: 02/04/2023]
Abstract
Recent technological advances have enabled massively parallel chromatin profiling with scATAC-seq (single-cell assay for transposase accessible chromatin by sequencing). Here we present ATAC with select antigen profiling by sequencing (ASAP-seq), a tool to simultaneously profile accessible chromatin and protein levels. Our approach pairs sparse scATAC-seq data with robust detection of hundreds of cell surface and intracellular protein markers and optional capture of mitochondrial DNA for clonal tracking, capturing three distinct modalities in single cells. ASAP-seq uses a bridging approach that repurposes antibody:oligonucleotide conjugates designed for existing technologies that pair protein measurements with single-cell RNA sequencing. Together with DOGMA-seq, an adaptation of CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing) for measuring gene activity across the central dogma of gene regulation, we demonstrate the utility of systematic multi-omic profiling by revealing coordinated and distinct changes in chromatin, RNA and surface proteins during native hematopoietic differentiation and peripheral blood mononuclear cell stimulation and as a combinatorial decoder and reporter of multiplexed perturbations in primary T cells.
Collapse
Affiliation(s)
- Eleni P Mimitou
- Technology Innovation Lab, New York Genome Center, New York, NY, USA
| | - Caleb A Lareau
- Department of Pathology, Stanford University, Stanford, CA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Hematology / Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kelvin Y Chen
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | | - Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Yusuke Takeshima
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Wendy Luo
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Hematology / Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | - Efthymia Papalexi
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | | | - Tatsuya Kibayashi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - James Badger Wing
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Human Immunology (Single Cell Immunology), Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Mayu Hata
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Rahul Satija
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | | | - Shimon Sakaguchi
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Leif S Ludwig
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Hematology / Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Vijay G Sankaran
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Hematology / Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- New York Genome Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Biology and Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Peter Smibert
- Technology Innovation Lab, New York Genome Center, New York, NY, USA.
| |
Collapse
|
155
|
Holthaus L, Sharma V, Brandt D, Ziegler AG, Jastroch M, Bonifacio E. Functional and metabolic fitness of human CD4 + T lymphocytes during metabolic stress. Life Sci Alliance 2021; 4:4/12/e202101013. [PMID: 34580176 PMCID: PMC8500231 DOI: 10.26508/lsa.202101013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Human T-cell activation, expansion, and effector function is grossly impaired in conditions that combine glucose deprivation and mild mitochondrial stress. Human CD4+ T cells are essential mediators of immune responses. By altering the mitochondrial and metabolic states, we defined metabolic requirements of human CD4+ T cells for in vitro activation, expansion, and effector function. T-cell activation and proliferation were reduced by inhibiting oxidative phosphorylation, whereas early cytokine production was maintained by either OXPHOS or glycolytic activity. Glucose deprivation in the presence of mild mitochondrial stress markedly reduced all three T-cell functions, contrasting the exposure to resveratrol, an antioxidant and sirtuin-1 activator, which specifically inhibited cytokine production and T-cell proliferation, but not T-cell activation. Conditions that inhibited T-cell activation were associated with the down-regulation of 2′,5′-oligoadenylate synthetase genes via interferon response pathways. Our findings indicate that T-cell function is grossly impaired by stressors combined with nutrient deprivation, suggesting that correcting nutrient availability, metabolic stress, and/or the function of T cells in these conditions will improve the efficacy of T-cell–based therapies.
Collapse
Affiliation(s)
- Lisa Holthaus
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Virag Sharma
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Daniel Brandt
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Martin Jastroch
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Ezio Bonifacio
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany .,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital and Faculty of Medicine of TU Dresden, Dresden, Germany
| |
Collapse
|
156
|
Harnessing Metabolic Reprogramming to Improve Cancer Immunotherapy. Int J Mol Sci 2021; 22:ijms221910268. [PMID: 34638609 PMCID: PMC8508898 DOI: 10.3390/ijms221910268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/31/2021] [Accepted: 09/04/2021] [Indexed: 02/06/2023] Open
Abstract
Immune escape is one of the hallmarks of cancer. While metabolic reprogramming provides survival advantage to tumor cancer cells, accumulating data also suggest such metabolic rewiring directly affects the activation, differentiation and function of immune cells, particularly in the tumor microenvironment. Understanding how metabolic reprogramming affects both tumor and immune cells, as well as their interplay, is therefore critical to better modulate tumor immune microenvironment in the era of cancer immunotherapy. In this review, we discuss alterations in several essential metabolic pathways in both tumor and key immune cells, provide evidence on their dynamic interaction, and propose innovative strategies to improve cancer immunotherapy via the modulation of metabolic pathways.
Collapse
|
157
|
Kuo CC, Chiang AWT, Baghdassarian HM, Lewis NE. Dysregulation of the secretory pathway connects Alzheimer's disease genetics to aggregate formation. Cell Syst 2021; 12:873-884.e4. [PMID: 34171228 PMCID: PMC8505362 DOI: 10.1016/j.cels.2021.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 02/24/2021] [Accepted: 06/02/2021] [Indexed: 12/14/2022]
Abstract
Amyloid disorders such as Alzheimer's disease (AD) involve the aggregation of secreted proteins. However, it is largely unclear how secretory-pathway proteins contribute to amyloid formation. We developed a systems biology framework integrating expression data with protein-protein interaction networks to estimate a tissue's fitness for producing specific secreted proteins and analyzed the fitness of the secretory pathway of various brain regions and cell types for synthesizing the AD-associated amyloid precursor protein (APP). While key amyloidogenic pathway components were not differentially expressed in AD brains, we found Aβ deposition correlates with systemic down- and upregulation of the secretory-pathway components proximal to APP and amyloidogenic secretases, respectively, in AD. Our analyses suggest that perturbations from three AD risk loci cascade through the APP secretory-support network and into the endocytosis pathway, connecting amyloidogenesis to dysregulation of secretory-pathway components supporting APP and suggesting novel therapeutic targets for AD. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Chih-Chung Kuo
- Department of Bioengineering, University of California, San Diego, San Diego, La Jolla, CA 92093, USA; Novo Nordisk Foundation Center for Biosustainability at UC San Diego, San Diego, La Jolla, CA 92093, USA
| | - Austin W T Chiang
- Novo Nordisk Foundation Center for Biosustainability at UC San Diego, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, San Diego, La Jolla, CA 92093, USA
| | - Hratch M Baghdassarian
- Department of Pediatrics, University of California, San Diego, San Diego, La Jolla, CA 92093, USA; Bioinformatics and Systems Biology Program, University of California, San Diego, San Diego, La Jolla, CA 92093, USA
| | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, San Diego, La Jolla, CA 92093, USA; Novo Nordisk Foundation Center for Biosustainability at UC San Diego, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
158
|
Jurgens AP, Popović B, Wolkers MC. T cells at work: How post-transcriptional mechanisms control T cell homeostasis and activation. Eur J Immunol 2021; 51:2178-2187. [PMID: 34180545 PMCID: PMC8457102 DOI: 10.1002/eji.202049055] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/07/2021] [Indexed: 12/19/2022]
Abstract
T cells are central players of the adaptive immune system by protecting us from recurring infections and by killing malignant cells. Protective T cell responses rely on the concerted production of effector molecules such as cytolytic mediators, granzymes, and perforins, as well as pro-inflammatory cytokines and chemokines. Once activated, T cells drastically change their gene expression and rapidly respond to insults by producing ample amounts of effector molecules. In the absence of antigen, T cells remain in a quiescent state and survey our body for possible pathogenic insults. Resting T cells are, however, not inert, but continuously regulate their protein production to survive and to be prepared for possible re-infections. Here, we review our current knowledge on the regulation of gene expression in activated and quiescent T cells. We specifically focus on post-transcriptional mechanisms that define the protein output and that allow dormant cells to undergo active signaling and selective translation, keeping them poised for activation. Finally, we discuss which signals drive T cell survival and their preparedness to respond to insults and which mechanisms are involved in these processes.
Collapse
Affiliation(s)
- Anouk P. Jurgens
- Department of HematopoiesisSanquin ResearchLandsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamOncode InstituteUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Branka Popović
- Department of HematopoiesisSanquin ResearchLandsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamOncode InstituteUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Monika C. Wolkers
- Department of HematopoiesisSanquin ResearchLandsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamOncode InstituteUtrechtThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| |
Collapse
|
159
|
Min K, Jannace TF, Si H, Veeramah KR, Haley JD, Konopka JB. Integrative multi-omics profiling reveals cAMP-independent mechanisms regulating hyphal morphogenesis in Candida albicans. PLoS Pathog 2021; 17:e1009861. [PMID: 34398936 PMCID: PMC8389844 DOI: 10.1371/journal.ppat.1009861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/26/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Microbial pathogens grow in a wide range of different morphologies that provide distinct advantages for virulence. In the fungal pathogen Candida albicans, adenylyl cyclase (Cyr1) is thought to be a master regulator of the switch to invasive hyphal morphogenesis and biofilm formation. However, faster growing cyr1Δ/Δ pseudorevertant (PR) mutants were identified that form hyphae in the absence of cAMP. Isolation of additional PR mutants revealed that their improved growth was due to loss of one copy of BCY1, the negative regulatory subunit of protein kinase A (PKA) from the left arm of chromosome 2. Furthermore, hyphal morphogenesis was improved in some of PR mutants by multigenic haploinsufficiency resulting from loss of large regions of the left arm of chromosome 2, including global transcriptional regulators. Interestingly, hyphal-associated genes were also induced in a manner that was independent of cAMP. This indicates that basal protein kinase A activity is an important prerequisite to induce hyphae, but activation of adenylyl cyclase is not needed. Instead, phosphoproteomic analysis indicated that the Cdc28 cyclin-dependent kinase and the casein kinase 1 family member Yck2 play key roles in promoting polarized growth. In addition, integrating transcriptomic and proteomic data reveals hyphal stimuli induce increased production of key transcription factors that contribute to polarized morphogenesis.
Collapse
Affiliation(s)
- Kyunghun Min
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University (SUNY), Stony Brook, New York, United States of America
| | - Thomas F. Jannace
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University (SUNY), Stony Brook, New York, United States of America
| | - Haoyu Si
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University (SUNY), Stony Brook, New York, United States of America
| | - Krishna R. Veeramah
- Department of Ecology and Evolution, Stony Brook University (SUNY), Stony Brook, New York, United States of America
| | - John D. Haley
- Department of Pathology, Renaissance School of Medicine, Stony Brook University (SUNY), Stony Brook, New York, United States of America
- Biological Mass Spectrometry Shared Resource, Renaissance School of Medicine, Stony Brook University (SUNY), Stony Brook, New York, United States of America
| | - James B. Konopka
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University (SUNY), Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
160
|
Diaz-Muñoz MD, Osma-Garcia IC. The RNA regulatory programs that govern lymphocyte development and function. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 13:e1683. [PMID: 34327847 DOI: 10.1002/wrna.1683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/25/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022]
Abstract
Lymphocytes require of constant and dynamic changes in their transcriptome for timely activation and production of effector molecules to combat external pathogens. Synthesis and translation of messenger (m)RNAs into these effector proteins is controlled both quantitatively and qualitatively by RNA binding proteins (RBPs). RBP-dependent regulation of RNA editing, subcellular location, stability, and translation shapes immune cell development and immunity. Extensive evidences have now been gathered from few model RBPs, HuR, PTBP1, ZFP36, and Roquin. However, recently developed methodologies for global characterization of protein:RNA interactions suggest the existence of complex RNA regulatory networks in which RBPs co-ordinately regulate the fate of sets of RNAs controlling cellular pathways and functions. In turn, RNA can also act as scaffolding of functionally related proteins modulating their activation and function. Here we review current knowledge about how RBP-dependent regulation of RNA shapes our immune system and discuss about the existence of a hidden immune cell epitranscriptome. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Manuel D Diaz-Muñoz
- Toulouse Institute for Infectious and Inflammatory Diseases, Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, Toulouse, France
| | - Ines C Osma-Garcia
- Toulouse Institute for Infectious and Inflammatory Diseases, Inserm UMR1291, CNRS UMR5051, University Paul Sabatier, Toulouse, France
| |
Collapse
|
161
|
Rodriguez-Coira J, Villaseñor A, Izquierdo E, Huang M, Barker-Tejeda TC, Radzikowska U, Sokolowska M, Barber D. The Importance of Metabolism for Immune Homeostasis in Allergic Diseases. Front Immunol 2021; 12:692004. [PMID: 34394086 PMCID: PMC8355700 DOI: 10.3389/fimmu.2021.692004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/05/2021] [Indexed: 12/27/2022] Open
Abstract
There is increasing evidence that the metabolic status of T cells and macrophages is associated with severe phenotypes of chronic inflammation, including allergic inflammation. Metabolic changes in immune cells have a crucial role in their inflammatory or regulatory responses. This notion is reinforced by metabolic diseases influencing global energy metabolism, such as diabetes or obesity, which are known risk factors of severity in inflammatory conditions, due to the metabolic-associated inflammation present in these patients. Since several metabolic pathways are closely tied to T cell and macrophage differentiation, a better understanding of metabolic alterations in immune disorders could help to restore and modulate immune cell functions. This link between energy metabolism and inflammation can be studied employing animal, human or cellular models. Analytical approaches rank from classic immunological studies to integrated analysis of metabolomics, transcriptomics, and proteomics. This review summarizes the main metabolic pathways of the cells involved in the allergic reaction with a focus on T cells and macrophages and describes different models and platforms of analysis used to study the immune system and its relationship with metabolism.
Collapse
Affiliation(s)
- Juan Rodriguez-Coira
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Alma Villaseñor
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Elena Izquierdo
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Tomás Clive Barker-Tejeda
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Domingo Barber
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| |
Collapse
|
162
|
Sun V, Sharpley M, Kaczor-Urbanowicz KE, Chang P, Montel-Hagen A, Lopez S, Zampieri A, Zhu Y, de Barros SC, Parekh C, Casero D, Banerjee U, Crooks GM. The Metabolic Landscape of Thymic T Cell Development In Vivo and In Vitro. Front Immunol 2021; 12:716661. [PMID: 34394122 PMCID: PMC8355594 DOI: 10.3389/fimmu.2021.716661] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/12/2021] [Indexed: 12/02/2022] Open
Abstract
Although metabolic pathways have been shown to control differentiation and activation in peripheral T cells, metabolic studies on thymic T cell development are still lacking, especially in human tissue. In this study, we use transcriptomics and extracellular flux analyses to investigate the metabolic profiles of primary thymic and in vitro-derived mouse and human thymocytes. Core metabolic pathways, specifically glycolysis and oxidative phosphorylation, undergo dramatic changes between the double-negative (DN), double-positive (DP), and mature single-positive (SP) stages in murine and human thymus. Remarkably, despite the absence of the complex multicellular thymic microenvironment, in vitro murine and human T cell development recapitulated the coordinated decrease in glycolytic and oxidative phosphorylation activity between the DN and DP stages seen in primary thymus. Moreover, by inducing in vitro T cell differentiation from Rag1-/- mouse bone marrow, we show that reduced metabolic activity at the DP stage is independent of TCR rearrangement. Thus, our findings suggest that highly conserved metabolic transitions are critical for thymic T cell development.
Collapse
Affiliation(s)
- Victoria Sun
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Molecular Biology Interdepartmental Program, UCLA, Los Angeles, CA, United States
| | - Mark Sharpley
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, CA, United States
| | - Karolina E. Kaczor-Urbanowicz
- Division of Oral Biology & Medicine, School of Dentistry, UCLA, Los Angeles, CA, United States
- Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, CA, United States
| | - Patrick Chang
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Molecular Biology Interdepartmental Program, UCLA, Los Angeles, CA, United States
| | - Amélie Montel-Hagen
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Shawn Lopez
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Alexandre Zampieri
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Yuhua Zhu
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Stéphanie C. de Barros
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Chintan Parekh
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - David Casero
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars- Sinai Medical Center, Los Angeles, CA, United States
| | - Utpal Banerjee
- Molecular Biology Interdepartmental Program, UCLA, Los Angeles, CA, United States
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, CA, United States
- Department of Biological Chemistry, UCLA, Los Angeles, CA, United States
- Eli and Edythe Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, United States
| | - Gay M. Crooks
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Eli and Edythe Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, United States
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| |
Collapse
|
163
|
Mayer KA, Smole U, Zhu C, Derdak S, Minervina AA, Salnikova M, Witzeneder N, Christamentl A, Boucheron N, Waidhofer-Söllner P, Trauner M, Hoermann G, Schmetterer KG, Mamedov IZ, Bilban M, Ellmeier W, Pickl WF, Gualdoni GA, Zlabinger GJ. The energy sensor AMPK orchestrates metabolic and translational adaptation in expanding T helper cells. FASEB J 2021; 35:e21217. [PMID: 33715236 PMCID: PMC8252394 DOI: 10.1096/fj.202001763rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 12/21/2022]
Abstract
The importance of cellular metabolic adaptation in inducing robust T cell responses is well established. However, the mechanism by which T cells link information regarding nutrient supply to clonal expansion and effector function is still enigmatic. Herein, we report that the metabolic sensor adenosine monophosphate-activated protein kinase (AMPK) is a critical link between cellular energy demand and translational activity and, thus, orchestrates optimal expansion of T cells in vivo. AMPK deficiency did not affect T cell fate decision, activation, or T effector cell generation; however, the magnitude of T cell responses in murine in vivo models of T cell activation was markedly reduced. This impairment was global, as all T helper cell subsets were similarly sensitive to loss of AMPK which resulted in reduced T cell accumulation in peripheral organs and reduced disease severity in pathophysiologically as diverse models as T cell transfer colitis and allergic airway inflammation. T cell receptor repertoire analysis confirmed similar clonotype frequencies in different lymphoid organs, thereby supporting the concept of a quantitative impairment in clonal expansion rather than a skewed qualitative immune response. In line with these findings, in-depth metabolic analysis revealed a decrease in T cell oxidative metabolism, and gene set enrichment analysis indicated a major reduction in ribosomal biogenesis and mRNA translation in AMPK-deficient T cells. We, thus, provide evidence that through its interference with these delicate processes, AMPK orchestrates the quantitative, but not the qualitative, manifestation of primary T cell responses in vivo.
Collapse
Affiliation(s)
- Katharina A Mayer
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ursula Smole
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ci Zhu
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Sophia Derdak
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Anastasia A Minervina
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Maria Salnikova
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Nadine Witzeneder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.,Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna Christamentl
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Nicole Boucheron
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Petra Waidhofer-Söllner
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.,MLL Munich Leukemia Laboratory, Munich, Germany
| | - Klaus G Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ilgar Z Mamedov
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Martin Bilban
- Core Facilities, Medical University of Vienna, Vienna, Austria.,Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Wilfried Ellmeier
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Guido A Gualdoni
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gerhard J Zlabinger
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
164
|
Zheng W, Wei J, Zebley CC, Jones LL, Dhungana Y, Wang YD, Mavuluri J, Long L, Fan Y, Youngblood B, Chi H, Geiger TL. Regnase-1 suppresses TCF-1+ precursor exhausted T-cell formation to limit CAR-T-cell responses against ALL. Blood 2021; 138:122-135. [PMID: 33690816 PMCID: PMC8288655 DOI: 10.1182/blood.2020009309] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapeutic efficacy is associated with long-term T-cell persistence and acquisition of memory. Memory-subset formation requires T-cell factor 1 (TCF-1), a master transcription factor for which few regulators have been identified. Here, we demonstrate using an immune-competent mouse model of B-cell acute lymphoblastic leukemia (ALL; B-ALL) that Regnase-1 deficiency promotes TCF-1 expression to enhance CAR-T-cell expansion and memory-like cell formation. This leads to improved CAR-T-mediated tumor clearance, sustained remissions, and protection against secondary tumor challenge. Phenotypic, transcriptional, and epigenetic profiling identified increased tumor-dependent programming of Regnase-1-deficient CAR-T cells into TCF-1+ precursor exhausted T cells (TPEX) characterized by upregulation of both memory and exhaustion markers. Regnase-1 directly targets Tcf7 messenger RNA (mRNA); its deficiency augments TCF-1 expression leading to the formation of TPEX that support long-term CAR-T-cell persistence and function. Regnase-1 deficiency also reduces exhaustion and enhances the activity of TCF-1- CAR-T cells. We further validate these findings in human CAR-T cells, where Regnase-1 deficiency mediates enhanced tumor clearance in a xenograft B-ALL model. This is associated with increased persistence and expansion of a TCF-1+ CAR-T-cell population. Our findings demonstrate the pivotal roles of TPEX, Regnase-1, and TCF-1 in mediating CAR-T-cell persistence and recall responses, and identify Regnase-1 as a modulator of human CAR-T-cell longevity and potency that may be manipulated for improved therapeutic efficacy.
Collapse
Affiliation(s)
- Wenting Zheng
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; and
| | - Jun Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; and
- Department of Immunology
| | | | - Lindsay L Jones
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | | | | | - Jayadev Mavuluri
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | | | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN
| | | | | | - Terrence L Geiger
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
165
|
Lee AM, Mansuri MS, Wilson RS, Lam TT, Nairn AC, Picciotto MR. Sex Differences in the Ventral Tegmental Area and Nucleus Accumbens Proteome at Baseline and Following Nicotine Exposure. Front Mol Neurosci 2021; 14:657064. [PMID: 34335180 PMCID: PMC8317211 DOI: 10.3389/fnmol.2021.657064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/09/2021] [Indexed: 12/20/2022] Open
Abstract
Sex differences in behaviors relevant to nicotine addiction have been observed in rodent models and human subjects. Behavioral, imaging, and epidemiological studies also suggest underlying sex differences in mesolimbic dopamine signaling pathways. In this study we evaluated the proteome in the ventral tegmental area (VTA) and nucleus accumbens (NAc) shell in male and female mice. Experimental groups included two mouse strains (C3H/HeJ and C57BL/6J) at baseline, a sub-chronic, rewarding regimen of nicotine in C3H/HeJ mice, and chronic nicotine administration and withdrawal in C57BL/6J mice. Isobaric labeling with a TMT 10-plex system, sample fractionation, and tandem mass spectrometry were used to quantify changes in protein abundance. In C3H/HeJ mice, similar numbers of proteins were differentially regulated between sexes at baseline compared with within each sex after sub-chronic nicotine administration. In C57BL/6J mice, there were significantly greater numbers of proteins differentially regulated between sexes at baseline compared with within each sex after chronic nicotine administration and withdrawal. Despite differences by sex, strain, and nicotine exposure parameters, glial fibrillary acidic protein (GFAP) and dopamine and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32, Ppp1r1b) were repeatedly identified as significantly altered proteins, especially in the VTA. Further, network analyses showed sex- and nicotine-dependent regulation of a number of signaling pathways, including dopaminergic signaling. Sub-chronic nicotine exposure in female mice increased proteins related to dopaminergic signaling in the NAc shell but decreased them in the VTA, whereas the opposite pattern was observed in male mice. In contrast, dopaminergic signaling pathways were similarly upregulated in both male and female VTA after chronic nicotine and withdrawal. Overall, this study identifies significant sex differences in the proteome of the mesolimbic system, at baseline and after nicotine reward or withdrawal, which may help explain differential trajectories and susceptibility to nicotine addiction in males and females.
Collapse
Affiliation(s)
- Angela M Lee
- Department of Psychiatry, Yale University, New Haven, CT, United States.,Yale Interdepartmental Neuroscience Program, New Haven, CT, United States
| | - Mohammad Shahid Mansuri
- Yale/NIDA Neuroproteomics Center, New Haven, CT, United States.,Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, United States
| | - Rashaun S Wilson
- Yale/NIDA Neuroproteomics Center, New Haven, CT, United States.,Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, United States.,W.M Keck Biotechnology Resource Laboratory, Yale University School of Medicine, New Haven, CT, United States
| | - TuKiet T Lam
- Yale/NIDA Neuroproteomics Center, New Haven, CT, United States.,Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, United States.,W.M Keck Biotechnology Resource Laboratory, Yale University School of Medicine, New Haven, CT, United States
| | - Angus C Nairn
- Department of Psychiatry, Yale University, New Haven, CT, United States.,Yale/NIDA Neuroproteomics Center, New Haven, CT, United States
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, New Haven, CT, United States.,Yale Interdepartmental Neuroscience Program, New Haven, CT, United States
| |
Collapse
|
166
|
IL-15 and PIM kinases direct the metabolic programming of intestinal intraepithelial lymphocytes. Nat Commun 2021; 12:4290. [PMID: 34257288 PMCID: PMC8277781 DOI: 10.1038/s41467-021-24473-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Intestinal intraepithelial lymphocytes (IEL) are an abundant population of tissue-resident T cells that protect and maintain the intestinal barrier. IEL respond to epithelial cell-derived IL-15, which is complexed to the IL-15 receptor α chain (IL-15/Rα). IL-15 is essential both for maintaining IEL homeostasis and inducing IEL responses to epithelial stress, which has been associated with Coeliac disease. Here, we apply quantitative mass spectrometry to IL-15/Rα-stimulated IEL to investigate how IL-15 directly regulates inflammatory functions of IEL. IL-15/Rα drives IEL activation through cell cycle regulation, upregulation of metabolic machinery and expression of a select repertoire of cell surface receptors. IL-15/Rα selectively upregulates the Ser/Thr kinases PIM1 and PIM2, which are essential for IEL to proliferate, grow and upregulate granzyme B in response to inflammatory IL-15. Notably, IEL from patients with Coeliac disease have high PIM expression. Together, these data indicate PIM kinases as important effectors of IEL responses to inflammatory IL-15.
Collapse
|
167
|
Galloway A, Kaskar A, Ditsova D, Atrih A, Yoshikawa H, Gomez-Moreira C, Suska O, Warminski M, Grzela R, Lamond AI, Darzynkiewicz E, Jemielity J, Cowling V. Upregulation of RNA cap methyltransferase RNMT drives ribosome biogenesis during T cell activation. Nucleic Acids Res 2021; 49:6722-6738. [PMID: 34125914 PMCID: PMC8266598 DOI: 10.1093/nar/gkab465] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/09/2021] [Accepted: 05/17/2021] [Indexed: 01/07/2023] Open
Abstract
The m7G cap is ubiquitous on RNAPII-transcribed RNA and has fundamental roles in eukaryotic gene expression, however its in vivo role in mammals has remained unknown. Here, we identified the m7G cap methyltransferase, RNMT, as a key mediator of T cell activation, which specifically regulates ribosome production. During T cell activation, induction of mRNA expression and ribosome biogenesis drives metabolic reprogramming, rapid proliferation and differentiation generating effector populations. We report that RNMT is induced by T cell receptor (TCR) stimulation and co-ordinates the mRNA, snoRNA and rRNA production required for ribosome biogenesis. Using transcriptomic and proteomic analyses, we demonstrate that RNMT selectively regulates the expression of terminal polypyrimidine tract (TOP) mRNAs, targets of the m7G-cap binding protein LARP1. The expression of LARP1 targets and snoRNAs involved in ribosome biogenesis is selectively compromised in Rnmt cKO CD4 T cells resulting in decreased ribosome synthesis, reduced translation rates and proliferation failure. By enhancing ribosome abundance, upregulation of RNMT co-ordinates mRNA capping and processing with increased translational capacity during T cell activation.
Collapse
Affiliation(s)
- Alison Galloway
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Aneesa Kaskar
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Dimitrinka Ditsova
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Abdelmadjid Atrih
- FingerPrints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Harunori Yoshikawa
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Carolina Gomez-Moreira
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Olga Suska
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Marcin Warminski
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Renata Grzela
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, and Division of Physics, 02-093 Warsaw, Poland
| | - Angus I Lamond
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Edward Darzynkiewicz
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, and Division of Physics, 02-093 Warsaw, Poland
| | - Jacek Jemielity
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Victoria H Cowling
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| |
Collapse
|
168
|
Zhou KL, Zhu ZH, Zhou JP, Zhao JJ, Zhang Y, Jiang B. Increased hexokinase-2 as a novel biomarker for the diagnosis and correlating with disease severity in rheumatoid arthritis. Medicine (Baltimore) 2021; 100:e26504. [PMID: 34160468 PMCID: PMC8238366 DOI: 10.1097/md.0000000000026504] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT Abnormal glucose metabolism brings out joint inflammation and destruction in rheumatoid arthritis (RA). The aim of this study was to evaluate the potential of circulating hexokinase-2 (HK2) in peripheral blood mononuclear cells (PBMCs) of rheumatoid arthritis (RA) patients.PBMCs were obtained from patients with RA or osteoarthritis (OA) and healthy controls (HCs). The expression of HK2 was assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The C-reactive protein (CRP) level, erythrocyte sedimentation rate (ESR), Calprotectin, rheumatoid factor (RF), anti-cyclic citrullinated peptides (anti-CCP) antibody level and 28-joint Disease Activity Score (DAS28), Clinical Disease Activity Index (CDAI) and Simplified Disease Activity Index (SDAI) were measured. Spearman's analysis was performed to determine the association between the level of HK2 and clinical characteristics. A receiver operating characteristic (ROC) curve was employed to evaluate the diagnostic value of HK2 in PBMCs. Logistic regression was used to identify risk factors. Sixty-five RA patients, 35 OA patients, and 40 HCs were included in the study.HK2 was upregulated in RA and OA patients compared with that in HCs (P < .05). The area under the ROC of HK2 for diagnosing RA and OA was 0.808 and 0.640, respectively. In addition, HK2 levels were increased in active RA compared with those in remittent RA (P = .03). Furthermore, HK2 correlated positively with the DAS28-ESR (P < .001), CDAI (P = .02) and SDAI scores (P = .02). Moreover, HK2 was independently associated with an increased risk of disease activity (DAS28-ESR>3.2, P = .02; CDAI score>10, P = .03; SDAI score>11, P = .04). Additionally, HK2 positivity was more frequently detected in patients treated with biologic disease-modifying antirheumatic drugs (bDMARDs) than in those not treated with bDMARDs.HK2 levels in PBMCs can be considered an ideal biomarker for diagnosing RA and involved in disease activity in RA. Dysregulation of HK2 may participate in the molecular mechanism of RA and could be an attractive selective metabolic target for RA treatment.
Collapse
|
169
|
Braun MY. The Natural History of T Cell Metabolism. Int J Mol Sci 2021; 22:ijms22136779. [PMID: 34202553 PMCID: PMC8269353 DOI: 10.3390/ijms22136779] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
The cells of the immune system, particularly the T lymphocytes, have two main features that distinguish them from the cells of other tissues. They proliferate after activation and have the ability to move in tissues and organs. These characteristics compel them to develop metabolic plasticity in order to fulfil their immune function. This review focuses on the different known mechanisms that allow T cells to adapt their metabolism to the real-life circumstances they operate in, whether it is to exit quiescence, to differentiate into effector cells, or to participate in immune memory formation. Some of the metabolic adaptations to environmental variations that T cells are likely to undergo in their immune monitoring function are also discussed.
Collapse
Affiliation(s)
- Michel Y Braun
- Institute for Medical Immunology (IMI), Faculty of Medicine, Université Libre de Bruxelles (ULB), 6041 Gosselies, Belgium
| |
Collapse
|
170
|
O'Sullivan D, Stanczak MA, Villa M, Uhl FM, Corrado M, Klein Geltink RI, Sanin DE, Apostolova P, Rana N, Edwards-Hicks J, Grzes KM, Kabat AM, Kyle RL, Fabri M, Curtis JD, Buck MD, Patterson AE, Regina A, Field CS, Baixauli F, Puleston DJ, Pearce EJ, Zeiser R, Pearce EL. Fever supports CD8 + effector T cell responses by promoting mitochondrial translation. Proc Natl Acad Sci U S A 2021; 118:e2023752118. [PMID: 34161266 PMCID: PMC8237659 DOI: 10.1073/pnas.2023752118] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Fever can provide a survival advantage during infection. Metabolic processes are sensitive to environmental conditions, but the effect of fever on T cell metabolism is not well characterized. We show that in activated CD8+ T cells, exposure to febrile temperature (39 °C) augmented metabolic activity and T cell effector functions, despite having a limited effect on proliferation or activation marker expression. Transcriptional profiling revealed an up-regulation of mitochondrial pathways, which was consistent with increased mass and metabolism observed in T cells exposed to 39 °C. Through in vitro and in vivo models, we determined that mitochondrial translation is integral to the enhanced metabolic activity and function of CD8+ T cells exposed to febrile temperature. Transiently exposing donor lymphocytes to 39 °C prior to infusion in a myeloid leukemia mouse model conferred enhanced therapeutic efficacy, raising the possibility that exposure of T cells to febrile temperatures could have clinical potential.
Collapse
Affiliation(s)
- David O'Sullivan
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Michal A Stanczak
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Matteo Villa
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Franziska M Uhl
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, 79106 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Mauro Corrado
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Ramon I Klein Geltink
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - David E Sanin
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Petya Apostolova
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Nisha Rana
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Joy Edwards-Hicks
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Katarzyna M Grzes
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Agnieszka M Kabat
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Ryan L Kyle
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Mario Fabri
- Department of Dermatology and Venereology, University of Cologne, 50937 Cologne, Germany
| | - Jonathan D Curtis
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Michael D Buck
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Annette E Patterson
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Annamaria Regina
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
- Department of Life Sciences, University of Trieste, 34128 Trieste, Italy
| | - Cameron S Field
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Francesc Baixauli
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Daniel J Puleston
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
| | - Edward J Pearce
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Erika L Pearce
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg im Breisgau, Germany;
| |
Collapse
|
171
|
Zhang W, Tang D, Lin L, Fan T, Xia L, Cai W, Dai W, Zou C, Yin L, Xu Y, Dai Y. Integrative multiplatform-based molecular profiling of human colorectal cancer reveals proteogenomic alterations underlying mitochondrial inactivation. Am J Cancer Res 2021; 11:2893-2910. [PMID: 34249434 PMCID: PMC8263689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/14/2021] [Indexed: 06/13/2023] Open
Abstract
Mitochondria play leading roles in initiation and progression of colorectal cancer (CRC). Proteogenomic analyses of mitochondria of CRC tumor cells would likely enhance our understanding of CRC pathogenesis and reveal new independent prognostic factors and treatment targets. However, comprehensive investigations focused on mitochondria of CRC patients are lacking. Here, we investigated global profiles of structural variants, DNA methylation, chromatin accessibility, transcriptome, proteome, and phosphoproteome on human CRC. Proteomic investigations uncovered greatly diminished mitochondrial proteome size in CRC relative to that found in adjacent healthy tissues. Integrated with analysis of RNA-Seq datasets obtained from the public database containing mRNA data of 538 CRC patients, the proteomic analysis indicated that proteins encoded by 45.5% of identified prognostic CRC genes were located within mitochondria, highlighting the association between altered mitochondrial function and CRC. Subsequently, we compared structural variants, DNA methylation, and chromatin accessibility of differentially expressed genes and found that chromatin accessibility was an important factor underlying mitochondrial gene expression. Furthermore, phosphoproteomic profiling demonstrated decreased phosphorylation of most mitochondria-related kinases within CRC versus adjacent healthy tissues, while also highlighting MKK3/p38 as an essential mitochondrial regulatory pathway. Meanwhile, systems-based analyses revealed identities of key kinases, transcriptional factors, and their interconnections. This research uncovered a close relationship between mitochondrial dysfunction and poor CRC prognosis, improve our understanding of molecular mechanism underlying mitochondrial linked to human CRC, and facilitate identifies of clinically relevant CRC prognostic factors and drug targets.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital)Shenzhen 518020, China
- The First Affiliated Hospital, Jinan UniversityGuangzhou, China
| | - Donge Tang
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital)Shenzhen 518020, China
| | - Liewen Lin
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital)Shenzhen 518020, China
| | - Tingting Fan
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua UniversityShenzhen 518055, China
| | - Ligang Xia
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital)Shenzhen 518020, China
| | - Wanxia Cai
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital)Shenzhen 518020, China
| | - Weier Dai
- College of Natural Science, University of Texas at AustinAustin 78721, United States of America
| | - Chang Zou
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital)Shenzhen 518020, China
| | - Lianghong Yin
- Department of Nephrology, Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan UniversityGuangzhou 510632, China
| | - Yong Xu
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s HospitalShenzhen 518028, China
| | - Yong Dai
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital)Shenzhen 518020, China
| |
Collapse
|
172
|
Raynor JL, Chapman NM, Chi H. Metabolic Control of Memory T-Cell Generation and Stemness. Cold Spring Harb Perspect Biol 2021; 13:cshperspect.a037770. [PMID: 33820774 DOI: 10.1101/cshperspect.a037770] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The formation of long-lived memory T cells is a critical feature of the adaptive immune response. T cells undergo metabolic reprogramming to establish a functional memory population. While initial studies characterized key metabolic pathways necessary for memory T-cell development, recent findings highlight that metabolic regulation of memory T-cell subsets is diverse. Here we describe the different requirements for metabolic programs and metabolism-related signaling pathways in memory T-cell development. We further discuss the contribution of cellular metabolism to memory T-cell functional reprogramming and stemness within acute and chronic inflammatory environments. Last, we highlight knowledge gaps and propose approaches to determine the roles of metabolites and metabolic enzymes in memory T-cell fate. Understanding how cellular metabolism regulates a functionally diverse memory population will undoubtedly provide new therapeutic insights to modulate protective T-cell immunity in human disease.
Collapse
Affiliation(s)
- Jana L Raynor
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| |
Collapse
|
173
|
Combinatory strategy using nanoscale proteomics and machine learning for T cell subtyping in peripheral blood of single multiple myeloma patients. Anal Chim Acta 2021; 1173:338672. [PMID: 34172147 DOI: 10.1016/j.aca.2021.338672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 11/23/2022]
Abstract
T cells play crucial roles in our immunity against hematological tumors by inducing sustained immune responses. Flow cytometry-based detection of a limited number of specific protein markers has been routinely applied for basic research and clinical investigation in this area. In this study, we combined flow cytometry with the simple integrated spintip-based proteomics technology (SISPROT) to characterize the proteome of primary T cell subtypes in the peripheral blood (PB) from single multiple myeloma (MM) patients. Taking advantage of the integrated high pH reversed-phase fractionation in the SISPROT device, the global proteomes of CD3+, CD4+ and CD8+ T cells were firstly profiled with a depth of >7 000 protein groups for each cell type. The sensitivity of single-shot proteomic analysis was dramatically improved by optimizing the SISPROT and data-dependent acquisition parameters for nanogram-level samples. Eight subtypes of T cells were sorted from about 4 mL PB of single MM patients, and the individual subtype-specific proteomes with coverage among 1 702 and 3 699 protein groups were obtained from as low as 70 ng and up to 500 ng of cell lysates. In addition, we developed a two-step machine learning-based subtyping strategy for proof-of-concept classifying eight T cell subtypes, independent of their cell numbers and individual differences. Our strategy demonstrates an easy-to-use proteomic analysis on immune cells with the potential to discover novel subtype-specific protein biomarkers from limited clinical samples in future large scale clinical studies.
Collapse
|
174
|
Schubert K, Karkossa I, Schor J, Engelmann B, Steinheuer LM, Bruns T, Rolle-Kampczyk U, Hackermüller J, von Bergen M. A Multi-Omics Analysis of Mucosal-Associated-Invariant T Cells Reveals Key Drivers of Distinct Modes of Activation. Front Immunol 2021; 12:616967. [PMID: 34108957 PMCID: PMC8183572 DOI: 10.3389/fimmu.2021.616967] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/19/2021] [Indexed: 12/17/2022] Open
Abstract
The function of mucosal-associated invariant T (MAIT) cells highly depends on the mode of activation, either by recognition of bacterial metabolites via their T cell receptor (TCR) or in a TCR-independent manner via cytokines. The underlying molecular mechanisms are not entirely understood. To define the activation of MAIT cells on the molecular level, we applied a multi-omics approach with untargeted transcriptomics, proteomics and metabolomics. Transcriptomic analysis of E. coli- and TCR-activated MAIT cells showed a distinct transcriptional reprogramming, including altered pathways, transcription factors and effector molecules. We validated the consequences of this reprogramming on the phenotype by proteomics and metabolomics. Thus, and to distinguish between TCR-dependent and -independent activation, MAIT cells were stimulated with IL12/IL18, anti-CD3/CD28 or both. Only a combination of both led to full activation of MAIT cells, comparable to activation by E. coli. Using an integrated network-based approach, we identified key drivers of the distinct modes of activation, including cytokines and transcription factors, as well as negative feedback regulators like TWIST1 or LAG3. Taken together, we present novel insights into the biological function of MAIT cells, which may represent a basis for therapeutic approaches to target MAIT cells in pathological conditions.
Collapse
Affiliation(s)
- Kristin Schubert
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Isabel Karkossa
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Jana Schor
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Beatrice Engelmann
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Lisa Maria Steinheuer
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Tony Bruns
- Department of Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule Aachen (RWTH), Aachen, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Jörg Hackermüller
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research (UFZ), Leipzig, Germany
- Institute of Biochemistry, Leipzig University, Leipzig, Germany
| |
Collapse
|
175
|
Fatangare A, Glässner A, Sachs B, Sickmann A. Future perspectives on in-vitro diagnosis of drug allergy by the lymphocyte transformation test. J Immunol Methods 2021; 495:113072. [PMID: 34000289 DOI: 10.1016/j.jim.2021.113072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/22/2021] [Accepted: 05/12/2021] [Indexed: 11/26/2022]
Abstract
This article aims to envisage future perspectives of the lymphocyte transformation test (LTT). We describe the select innovative techniques, which can be integrated at different stages of the LTT to potentially improve the sensitivity, specificity, or practicability of the LTT. We first focus upon the cell sorting techniques comprising immunomagnetic cell separation and flow cytometry, which can be implemented prior and after the LTT culturing step to concentrate and quantify specific immune cell types. Further, we elaborate upon three important omics techniques such as transcriptomics, proteomics, and metabolomics, which can be integrated downstream of the LTT to analyze molecular changes in specific immune cells following drug induced activation and proliferation. We also develop visions, how state of the art techniques used in other scientific fields, can be transferred and applied in the context of in-vitro detection of drug allergy.
Collapse
Affiliation(s)
- Amol Fatangare
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Straße 11, 44139 Dortmund, Germany
| | - Andreas Glässner
- Federal Institute for Drugs and Medical Devices, Research Division, Bonn, Germany
| | - Bernhardt Sachs
- Federal Institute for Drugs and Medical Devices, Research Division, Bonn, Germany; Department for Dermatology and Allergology, University Hospital Aachen, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Straße 11, 44139 Dortmund, Germany; Medizinische Fakultät, Medizinisches Proteom-Center (MPC), Ruhr-Universität Bochum, 44801 Bochum, Germany; Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, AB243FX, Scotland, UK.
| |
Collapse
|
176
|
Renoux F, Stellato M, Haftmann C, Vogetseder A, Huang R, Subramaniam A, Becker MO, Blyszczuk P, Becher B, Distler JHW, Kania G, Boyman O, Distler O. The AP1 Transcription Factor Fosl2 Promotes Systemic Autoimmunity and Inflammation by Repressing Treg Development. Cell Rep 2021; 31:107826. [PMID: 32610127 DOI: 10.1016/j.celrep.2020.107826] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/27/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
Regulatory T cells (Tregs) represent a major population in the control of immune homeostasis and autoimmunity. Here we show that Fos-like 2 (Fosl2), a TCR-induced AP1 transcription factor, represses Treg development and controls autoimmunity. Mice overexpressing Fosl2 (Fosl2tg) indeed show a systemic inflammatory phenotype, with immune infiltrates in multiple organs. This phenotype is absent in Fosl2tg × Rag2-/- mice lacking T and B cells, and Fosl2 induces T cell-intrinsic reduction of Treg development that is responsible for the inflammatory phenotype. Fosl2tg T cells can transfer inflammation, which is suppressed by the co-delivery of Tregs, while Fosl2 deficiency in T cells reduces the severity of autoimmunity in the EAE model. We find that Fosl2 could affect expression of FoxP3 and other Treg development genes. Our data highlight the importance of AP1 transcription factors, in particular Fosl2, during T cell development to determine Treg differentiation and control autoimmunity.
Collapse
Affiliation(s)
- Florian Renoux
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Mara Stellato
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Claudia Haftmann
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Riyun Huang
- Sanofi, Immunology and Inflammation Research TA, Cambridge, MA 02139, USA
| | - Arun Subramaniam
- Sanofi, Immunology and Inflammation Research TA, Cambridge, MA 02139, USA
| | - Mike O Becker
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Przemyslaw Blyszczuk
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland; Department of Clinical Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jörg H W Distler
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Gabriela Kania
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
177
|
Zheng Z, Zhang L, Cui XL, Yu X, Hsu PJ, Lyu R, Tan H, Mandal M, Zhang M, Sun HL, Sanchez Castillo A, Peng J, Clark MR, He C, Huang H. Control of Early B Cell Development by the RNA N 6-Methyladenosine Methylation. Cell Rep 2021; 31:107819. [PMID: 32610122 DOI: 10.1016/j.celrep.2020.107819] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/18/2020] [Accepted: 06/03/2020] [Indexed: 12/21/2022] Open
Abstract
The RNA N6-methyladenosine (m6A) methylation is installed by the METTL3-METTL14 methyltransferase complex. This modification has critical regulatory roles in various biological processes. Here, we report that deletion of Mettl14 dramatically reduces mRNA m6A methylation in developing B cells and severely blocks B cell development in mice. Deletion of Mettl14 impairs interleukin-7 (IL-7)-induced pro-B cell proliferation and the large-pre-B-to-small-pre-B transition and causes dramatic abnormalities in gene expression programs important for B cell development. Suppression of a group of transcripts by cytoplasmic m6A reader YTHDF2 is critical to the IL-7-induced pro-B cell proliferation. In contrast, the block in the large-pre-B-to-small-pre-B transition is independent of YTHDF1 or YTHDF2 but is associated with a failure to properly upregulate key transcription factors regulating this transition. Our data highlight the important regulatory roles of the RNA m6A methylation and its reader proteins in early B cell development.
Collapse
Affiliation(s)
- Zhong Zheng
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA; Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL 60637, USA; Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Linda Zhang
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA; Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Xiao-Long Cui
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Xianbin Yu
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Phillip J Hsu
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA
| | - Ruitu Lyu
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Haiyan Tan
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Malay Mandal
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL 60637, USA
| | - Michelle Zhang
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA; Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL 60637, USA
| | - Hui-Lung Sun
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Arantxa Sanchez Castillo
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Marcus R Clark
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA
| | - Chuan He
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA; Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA.
| | - Haochu Huang
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
178
|
Abstract
Recent evidence supports the notion that mitochondrial metabolism is necessary for T cell activation, proliferation, and function. Mitochondrial metabolism supports T cell anabolism by providing key metabolites for macromolecule synthesis and generating metabolites for T cell function. In this review, we focus on how mitochondrial metabolism controls conventional and regulatory T cell fates and function.
Collapse
Affiliation(s)
- Elizabeth M Steinert
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Karthik Vasan
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Navdeep S Chandel
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| |
Collapse
|
179
|
Extracellular signal-regulated kinase (ERK) pathway control of CD8+ T cell differentiation. Biochem J 2021; 478:79-98. [PMID: 33305809 PMCID: PMC7813476 DOI: 10.1042/bcj20200661] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 01/14/2023]
Abstract
The integration of multiple signalling pathways that co-ordinate T cell metabolism and transcriptional reprogramming is required to drive T cell differentiation and proliferation. One key T cell signalling module is mediated by extracellular signal-regulated kinases (ERKs) which are activated in response to antigen receptor engagement. The activity of ERKs is often used to report antigen receptor occupancy but the full details of how ERKs control T cell activation is not understood. Accordingly, we have used mass spectrometry to explore how ERK signalling pathways control antigen receptor driven proteome restructuring in CD8+ T cells to gain insights about the biological processes controlled by ERKs in primary lymphocytes. Quantitative analysis of >8000 proteins identified 900 ERK regulated proteins in activated CD8+ T cells. The data identify both positive and negative regulatory roles for ERKs during T cell activation and reveal that ERK signalling primarily controls the repertoire of transcription factors, cytokines and cytokine receptors expressed by activated T cells. It was striking that a large proportion of the proteome restructuring that is driven by triggering of the T cell antigen receptor is not dependent on ERK activation. However, the selective targets of the ERK signalling module include the critical effector molecules and the cytokines that allow T cell communication with other immune cells to mediate adaptive immune responses.
Collapse
|
180
|
Rossi A, Pacella I, Piconese S. RNA Flow Cytometry for the Study of T Cell Metabolism. Int J Mol Sci 2021; 22:ijms22083906. [PMID: 33918901 PMCID: PMC8069477 DOI: 10.3390/ijms22083906] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/30/2022] Open
Abstract
T cells undergo activation and differentiation programs along a continuum of states that can be tracked through flow cytometry using a combination of surface and intracellular markers. Such dynamic behavior is the result of transcriptional and post-transcriptional events, initiated and sustained by the activation of specific transcription factors and by epigenetic remodeling. These signaling pathways are tightly integrated with metabolic routes in a bidirectional manner: on the one hand, T cell receptors and costimulatory molecules activate metabolic reprogramming; on the other hand, metabolites modify T cell transcriptional programs and functions. Flow cytometry represents an invaluable tool to analyze the integration of phenotypical, functional, metabolic and transcriptional features, at the single cell level in heterogeneous T cell populations, and from complex microenvironments, with potential clinical application in monitoring the efficacy of cancer immunotherapy. Here, we review the most recent advances in flow cytometry-based analysis of gene expression, in combination with indicators of mitochondrial activity, with the aim of revealing and characterizing major metabolic pathways in T cells.
Collapse
Affiliation(s)
- Alessandra Rossi
- Department of Internal Clinical Sciences, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Roma, Italy; (A.R.); (I.P.)
| | - Ilenia Pacella
- Department of Internal Clinical Sciences, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Roma, Italy; (A.R.); (I.P.)
| | - Silvia Piconese
- Department of Internal Clinical Sciences, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Roma, Italy; (A.R.); (I.P.)
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Roma, Italy
- Correspondence:
| |
Collapse
|
181
|
Papadopoulou G, Xanthou G. Metabolic rewiring: a new master of Th17 cell plasticity and heterogeneity. FEBS J 2021; 289:2448-2466. [PMID: 33794075 DOI: 10.1111/febs.15853] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/21/2021] [Accepted: 03/29/2021] [Indexed: 01/04/2023]
Abstract
T helper type 17 (Th17) cells are characterized by inherent plasticity and heterogeneity displaying both pathogenic and tissue-protective functions. Emerging evidence has illuminated a pivotal role for metabolic reprogramming in shaping Th17 cell fate determination. Metabolic responses are regulated by a constellation of factors and environmental triggers, including cytokines, nutrients, oxygen levels, and metabolites. Dysregulation of metabolic pathways not only influences Th17 cell plasticity and effector function but also affects the outcome of Th17-linked autoimmune, inflammatory, and antitumor responses. Understanding the molecular mechanisms underpinning metabolic reprogramming can allow the enhancement of protective Th17 cell-mediated responses during infections and cancer, concomitant with the suppression of detrimental Th17 processes during autoimmune and inflammatory diseases. In the present review, we describe major metabolic pathways underlying the differentiation of Th17 cells and their crosstalk with intracellular signaling mediators, we discuss how metabolic reprogramming affects Th17 cell plasticity and functions, and, finally, we outline current advances in the exploitation of metabolic checkpoints for the development of novel therapeutic interventions for the management of tissue inflammation, autoimmune disorders, and cancer.
Collapse
Affiliation(s)
- Gina Papadopoulou
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Greece.,Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Greece
| | - Georgina Xanthou
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
182
|
Paxbp1 controls a key checkpoint for cell growth and survival during early activation of quiescent muscle satellite cells. Proc Natl Acad Sci U S A 2021; 118:2021093118. [PMID: 33753492 DOI: 10.1073/pnas.2021093118] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Adult mouse muscle satellite cells (MuSCs) are quiescent in uninjured muscles. Upon muscle injury, MuSCs exit quiescence, reenter the cell cycle to proliferate and self-renew, and then differentiate and fuse to drive muscle regeneration. However, it remains poorly understood how MuSCs transition from quiescence to the cycling state. Here, we report that Pax3 and Pax7 binding protein 1 (Paxbp1) controls a key checkpoint during this critical transition. Deletion of Paxbp1 in adult MuSCs prevented them from reentering the cell cycle upon injury, resulting in a total regeneration failure. Mechanistically, we found an abnormal elevation of reactive oxygen species (ROS) in Paxbp1-null MuSCs, which induced p53 activation and impaired mTORC1 signaling, leading to defective cell growth, apoptosis, and failure in S-phase reentry. Deliberate ROS reduction partially rescued the cell-cycle reentry defect in mutant MuSCs. Our study reveals that Paxbp1 regulates a late cell-growth checkpoint essential for quiescent MuSCs to reenter the cell cycle upon activation.
Collapse
|
183
|
T cells: a dedicated effector kinase pathways for every trait? Biochem J 2021; 478:1303-1307. [PMID: 33755101 DOI: 10.1042/bcj20210006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 11/17/2022]
Abstract
Signaling pathways play critical roles in regulating the activation of T cells. Recognition of foreign peptide presented by MHC to the T cell receptor (TCR) triggers a signaling cascade of proximal kinases and adapter molecules that lead to the activation of Effector kinase pathways. These effector kinase pathways play pivotal roles in T cell activation, differentiation, and proliferation. RNA sequencing-based methods have provided insights into the gene expression programs that support the above-mentioned cell biological responses. The proteome is often overlooked. A recent study by Damasio et al. [Biochem. J. (2021) 478, 79-98. doi:10.1042/BCJ20200661] focuses on characterizing the effect of extracellular signal-regulated kinase (ERK) on the remodeling of the proteome of activated CD8+ T cells using Mass spectrometric analysis. Surprisingly, the Effector kinase ERK pathway is responsible for only a select proportion of the proteome that restructures during T cell activation. The primary targets of ERK signaling are transcription factors, cytokines, and cytokine receptors. In this commentary, we discuss the recent findings by Damasio et al. [Biochem. J. (2021) 478, 79-98. doi:10.1042/BCJ20200661] in the context of different Effector kinase pathways in activated T cells.
Collapse
|
184
|
Rad S. M. AH, Halpin JC, Mollaei M, Smith Bell SWJ, Hirankarn N, McLellan AD. Metabolic and Mitochondrial Functioning in Chimeric Antigen Receptor (CAR)-T Cells. Cancers (Basel) 2021; 13:1229. [PMID: 33799768 PMCID: PMC8002030 DOI: 10.3390/cancers13061229] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 02/02/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized adoptive cell therapy with impressive therapeutic outcomes of >80% complete remission (CR) rates in some haematological malignancies. Despite this, CAR T cell therapy for the treatment of solid tumours has invariably been unsuccessful in the clinic. Immunosuppressive factors and metabolic stresses in the tumour microenvironment (TME) result in the dysfunction and exhaustion of CAR T cells. A growing body of evidence demonstrates the importance of the mitochondrial and metabolic state of CAR T cells prior to infusion into patients. The different T cell subtypes utilise distinct metabolic pathways to fulfil their energy demands associated with their function. The reprogramming of CAR T cell metabolism is a viable approach to manufacture CAR T cells with superior antitumour functions and increased longevity, whilst also facilitating their adaptation to the nutrient restricted TME. This review discusses the mitochondrial and metabolic state of T cells, and describes the potential of the latest metabolic interventions to maximise CAR T cell efficacy for solid tumours.
Collapse
Affiliation(s)
- Ali Hosseini Rad S. M.
- Department of Microbiology and Immunology, University of Otago, Dunedin 9010, Otago, New Zealand; (J.C.H.); (S.W.J.S.B.)
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok 10330, Thailand
| | - Joshua Colin Halpin
- Department of Microbiology and Immunology, University of Otago, Dunedin 9010, Otago, New Zealand; (J.C.H.); (S.W.J.S.B.)
| | - Mojtaba Mollaei
- Department of Immunology, School of Medicine, Tarbiat Modares University, Tehran 14117-13116, Iran;
| | - Samuel W. J. Smith Bell
- Department of Microbiology and Immunology, University of Otago, Dunedin 9010, Otago, New Zealand; (J.C.H.); (S.W.J.S.B.)
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok 10330, Thailand
| | - Alexander D. McLellan
- Department of Microbiology and Immunology, University of Otago, Dunedin 9010, Otago, New Zealand; (J.C.H.); (S.W.J.S.B.)
| |
Collapse
|
185
|
Lemaitre P, Bai Q, Legrand C, Chariot A, Close P, Bureau F, Desmet CJ. Loss of the Transfer RNA Wobble Uridine-Modifying Enzyme Elp3 Delays T Cell Cycle Entry and Impairs T Follicular Helper Cell Responses through Deregulation of Atf4. THE JOURNAL OF IMMUNOLOGY 2021; 206:1077-1087. [PMID: 33483347 DOI: 10.4049/jimmunol.2000521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/19/2020] [Indexed: 12/24/2022]
Abstract
The activation of T cells is accompanied by intensive posttranscriptional remodeling of their proteome. We observed that protein expression of enzymes that modify wobble uridine in specific tRNAs, namely elongator subunit 3 (Elp3) and cytosolic thiouridylase (Ctu)2, increased in the course of T cell activation. To investigate the role of these tRNA epitranscriptomic modifiers in T cell biology, we generated mice deficient for Elp3 in T cells. We show that deletion of Elp3 has discrete effects on T cells. In vitro, Elp3-deficient naive CD4+ T cells polarize normally but are delayed in entering the first cell cycle following activation. In vivo, different models of immunization revealed that Elp3-deficient T cells display reduced expansion, resulting in functional impairment of T follicular helper (TFH) responses, but not of other CD4+ effector T cell responses. Transcriptomic analyses identified a progressive overactivation of the stress-responsive transcription factor Atf4 in Elp3-deficient T cells. Overexpression of Atf4 in wild-type T cells phenocopies the effect of Elp3 loss on T cell cycle entry and TFH cell responses. Reciprocally, partial silencing of Atf4 or deletion of its downstream effector transcription factor Chop rescues TFH responses of Elp3-deficient T cells. Together, our results reveal that specific epitranscriptomic tRNA modifications contribute to T cell cycle entry and promote optimal TFH responses.
Collapse
Affiliation(s)
- Pierre Lemaitre
- Laboratory of Cellular and Molecular Immunology, Interdisciplinary Group for Applied Genoproteomics, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Qiang Bai
- Laboratory of Cellular and Molecular Immunology, Interdisciplinary Group for Applied Genoproteomics, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Céline Legrand
- Laboratory of Cellular and Molecular Immunology, Interdisciplinary Group for Applied Genoproteomics, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium
| | - Alain Chariot
- Laboratory of Medical Chemistry, Interdisciplinary Group for Applied Genoproteomics, Faculty of Medicine, University of Liege, 4000 Liege, Belgium.,Walloon Excellence in Life Sciences and Biotechnology, 1300 Wavres, Belgium; and
| | - Pierre Close
- Walloon Excellence in Life Sciences and Biotechnology, 1300 Wavres, Belgium; and.,Laboratory of Cancer Signaling, Interdisciplinary Group for Applied Genoproteomics, Faculty of Medicine, University of Liege, 4000 Liege, Belgium
| | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, Interdisciplinary Group for Applied Genoproteomics, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium.,Walloon Excellence in Life Sciences and Biotechnology, 1300 Wavres, Belgium; and
| | - Christophe J Desmet
- Laboratory of Cellular and Molecular Immunology, Interdisciplinary Group for Applied Genoproteomics, Faculty of Veterinary Medicine, University of Liege, 4000 Liege, Belgium; .,Walloon Excellence in Life Sciences and Biotechnology, 1300 Wavres, Belgium; and
| |
Collapse
|
186
|
Curdy N, Lanvin O, Cadot S, Laurent C, Fournié JJ, Franchini DM. Stress Granules in the Post-transcriptional Regulation of Immune Cells. Front Cell Dev Biol 2021; 8:611185. [PMID: 33520991 PMCID: PMC7841200 DOI: 10.3389/fcell.2020.611185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
Immune cell activation triggers transcriptional and translational programs eliciting cellular processes, such as differentiation or proliferation, essential for an efficient immune response. These dynamic processes require an intricate orchestration of regulatory mechanisms to control the precise spatiotemporal expression of proteins. Post-transcriptional regulation ensures the control of messenger RNA metabolism and appropriate translation. Among these post-transcriptional regulatory mechanisms, stress granules participate in the control of protein synthesis. Stress granules are ribonucleoprotein complexes that form upon stress, typically under control of the integrated stress response. Such structures assemble upon stimulation of immune cells where they control selective translational programs ensuring the establishment of accurate effector functions. In this review, we summarize the current knowledge about post-transcriptional regulation in immune cells and highlight the role of stress sensors and stress granules in such regulation.
Collapse
Affiliation(s)
- Nicolas Curdy
- Cancer Research Center of Toulouse (CRCT), INSERM UMR 1037, CNRS ERL 5294, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Olivia Lanvin
- Cancer Research Center of Toulouse (CRCT), INSERM UMR 1037, CNRS ERL 5294, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Sarah Cadot
- Cancer Research Center of Toulouse (CRCT), INSERM UMR 1037, CNRS ERL 5294, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Camille Laurent
- Cancer Research Center of Toulouse (CRCT), INSERM UMR 1037, CNRS ERL 5294, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France.,Département de Pathologie, Centre Hospitalier Universitaire (CHU) de Toulouse, Toulouse, France
| | - Jean-Jacques Fournié
- Cancer Research Center of Toulouse (CRCT), INSERM UMR 1037, CNRS ERL 5294, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Don-Marc Franchini
- Cancer Research Center of Toulouse (CRCT), INSERM UMR 1037, CNRS ERL 5294, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| |
Collapse
|
187
|
Subbannayya Y, Haug M, Pinto SM, Mohanty V, Meås HZ, Flo TH, Prasad TK, Kandasamy RK. The Proteomic Landscape of Resting and Activated CD4+ T Cells Reveal Insights into Cell Differentiation and Function. Int J Mol Sci 2020; 22:E275. [PMID: 33383959 PMCID: PMC7795831 DOI: 10.3390/ijms22010275] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/17/2020] [Accepted: 12/24/2020] [Indexed: 12/14/2022] Open
Abstract
CD4+ T cells (T helper cells) are cytokine-producing adaptive immune cells that activate or regulate the responses of various immune cells. The activation and functional status of CD4+ T cells is important for adequate responses to pathogen infections but has also been associated with auto-immune disorders and survival in several cancers. In the current study, we carried out a label-free high-resolution FTMS-based proteomic profiling of resting and T cell receptor-activated (72 h) primary human CD4+ T cells from peripheral blood of healthy donors as well as SUP-T1 cells. We identified 5237 proteins, of which significant alterations in the levels of 1119 proteins were observed between resting and activated CD4+ T cells. In addition to identifying several known T-cell activation-related processes altered expression of several stimulatory/inhibitory immune checkpoint markers between resting and activated CD4+ T cells were observed. Network analysis further revealed several known and novel regulatory hubs of CD4+ T cell activation, including IFNG, IRF1, FOXP3, AURKA, and RIOK2. Comparison of primary CD4+ T cell proteomic profiles with human lymphoblastic cell lines revealed a substantial overlap, while comparison with mouse CD+ T cell data suggested interspecies proteomic differences. The current dataset will serve as a valuable resource to the scientific community to compare and analyze the CD4+ proteome.
Collapse
Affiliation(s)
- Yashwanth Subbannayya
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway; (Y.S.); (M.H.); (S.M.P.); (H.Z.M.); (T.H.F.)
| | - Markus Haug
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway; (Y.S.); (M.H.); (S.M.P.); (H.Z.M.); (T.H.F.)
| | - Sneha M. Pinto
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway; (Y.S.); (M.H.); (S.M.P.); (H.Z.M.); (T.H.F.)
| | - Varshasnata Mohanty
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, India; (V.M.); (T.S.K.P.)
| | - Hany Zakaria Meås
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway; (Y.S.); (M.H.); (S.M.P.); (H.Z.M.); (T.H.F.)
| | - Trude Helen Flo
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway; (Y.S.); (M.H.); (S.M.P.); (H.Z.M.); (T.H.F.)
| | - T.S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, India; (V.M.); (T.S.K.P.)
| | - Richard K. Kandasamy
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway; (Y.S.); (M.H.); (S.M.P.); (H.Z.M.); (T.H.F.)
| |
Collapse
|
188
|
Yukl SA, Khan S, Chen TH, Trapecar M, Wu F, Xie G, Telwatte S, Fulop D, Pico AR, Laird GM, Ritter KD, Jones NG, Lu CM, Siliciano RF, Roan NR, Milush JM, Somsouk M, Deeks SG, Hunt PW, Sanjabi S. Shared Mechanisms Govern HIV Transcriptional Suppression in Circulating CD103 + and Gut CD4 + T Cells. J Virol 2020; 95:e01331-20. [PMID: 33115867 PMCID: PMC7944458 DOI: 10.1128/jvi.01331-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/23/2020] [Indexed: 12/22/2022] Open
Abstract
Latent HIV infection is the main barrier to cure, and most HIV-infected cells reside in the gut, where distinct but unknown mechanisms may promote viral latency. Transforming growth factor β (TGF-β), which induces the expression of CD103 on tissue-resident memory T cells, has been implicated in HIV latency. Using CD103 as a surrogate marker to identify cells that have undergone TGF-β signaling, we compared the HIV RNA/DNA contents and cellular transcriptomes of CD103+ and CD103- CD4 T cells from the blood and rectum of HIV-negative (HIV-) and antiretroviral therapy (ART)-suppressed HIV-positive (HIV+) individuals. Like gut CD4+ T cells, circulating CD103+ cells harbored more HIV DNA than did CD103- cells but transcribed less HIV RNA per provirus. Circulating CD103+ cells also shared a gene expression profile that is closer to that of gut CD4 T cells than to that of circulating CD103- cells, with significantly lower expression levels of ribosomal proteins and transcriptional and translational pathways associated with HIV expression but higher expression levels of a subset of genes implicated in suppressing HIV transcription. These findings suggest that blood CD103+ CD4 T cells can serve as a model to study the molecular mechanisms of HIV latency in the gut and reveal new cellular factors that may contribute to HIV latency.IMPORTANCE The ability of HIV to establish a reversibly silent, "latent" infection is widely regarded as the main barrier to curing HIV. Most HIV-infected cells reside in tissues such as the gut, but it is unclear what mechanisms maintain HIV latency in the blood or gut. We found that circulating CD103+ CD4+ T cells are enriched for HIV-infected cells in a latent-like state. Using RNA sequencing (RNA-seq), we found that CD103+ T cells share a cellular transcriptome that more closely resembles that of CD4+ T cells from the gut, suggesting that they are homing to or from the gut. We also identified the cellular genes whose expression distinguishes gut CD4+ or circulating CD103+ T cells from circulating CD103- T cells, including some genes that have been implicated in HIV expression. These genes may contribute to latent HIV infection in the gut and may serve as new targets for therapies aimed at curing HIV.
Collapse
Affiliation(s)
- Steven A Yukl
- San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, California, USA
| | - Shahzada Khan
- Gladstone Institutes, San Francisco, California, USA
| | - Tsui-Hua Chen
- San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
- University of California, San Francisco, San Francisco, California, USA
| | | | - Frank Wu
- Gladstone Institutes, San Francisco, California, USA
| | - Guorui Xie
- Gladstone Institutes, San Francisco, California, USA
- Department of Urology, University of California, San Francisco, San Francisco, California, USA
| | - Sushama Telwatte
- San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, California, USA
| | - Daniel Fulop
- Gladstone Institutes, San Francisco, California, USA
| | | | | | | | - Norman G Jones
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, California, USA
| | - Chuanyi M Lu
- San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
- University of California, San Francisco, San Francisco, California, USA
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Howard Hughes Medical Institute, Baltimore, Maryland, USA
| | - Nadia R Roan
- Gladstone Institutes, San Francisco, California, USA
- Department of Urology, University of California, San Francisco, San Francisco, California, USA
| | - Jeffrey M Milush
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, California, USA
| | - Ma Somsouk
- Division of Gastroenterology, University of California, San Francisco, San Francisco, California, USA
| | - Steven G Deeks
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, California, USA
| | - Peter W Hunt
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, California, USA
| | - Shomyseh Sanjabi
- Gladstone Institutes, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
189
|
Protein Prenylation Drives Discrete Signaling Programs for the Differentiation and Maintenance of Effector T reg Cells. Cell Metab 2020; 32:996-1011.e7. [PMID: 33207246 PMCID: PMC7887758 DOI: 10.1016/j.cmet.2020.10.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 08/09/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
Effector regulatory T (eTreg) cells are essential for immune tolerance and depend upon T cell receptor (TCR) signals for generation. The immunometabolic signaling mechanisms that promote the differentiation and maintenance of eTreg cells remain unclear. Here, we show that isoprenoid-dependent posttranslational lipid modifications dictate eTreg cell accumulation and function by intersecting with TCR-induced intracellular signaling. We find that isoprenoids are essential for activated Treg cell suppressive activity, and Treg cell-specific deletion of the respective farnesylation- and geranylgeranylation-promoting enzymes Fntb or Pggt1b leads to the development of fatal autoimmunity, associated with reduced eTreg cell accumulation. Mechanistically, Fntb promotes eTreg cell maintenance by regulating mTORC1 activity and ICOS expression. In contrast, Pggt1b acts as a rheostat of TCR-dependent transcriptional programming and Rac-mediated signaling for establishment of eTreg cell differentiation and immune tolerance. Therefore, our results identify bidirectional metabolic signaling, specifically between immunoreceptor signaling and metabolism-mediated posttranslational lipid modifications, for the differentiation and maintenance of eTreg cells.
Collapse
|
190
|
Barili V, Boni C, Rossi M, Vecchi A, Zecca A, Penna A, Missale G, Ferrari C, Fisicaro P. Metabolic regulation of the HBV-specific T cell function. Antiviral Res 2020; 185:104989. [PMID: 33248194 DOI: 10.1016/j.antiviral.2020.104989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/15/2022]
Abstract
Chronically HBV infected subjects are more than 260 million worldwide; cirrhosis and liver cancer represent possible outcomes which affect around 700,000 patients per year. Both innate and adaptive immune responses are necessary for viral control and both have been shown to be defective in chronic patients. Metabolic remodeling is an essential process in T cell biology, particularly for T cell activation, differentiation and survival. Cellular metabolism relies on the conversion of nutrients into energy to support intracellular processes, and to generate fundamental intermediate components for cell proliferation and growth. Adaptive immune responses are the central mechanisms for the resolution of primary human infections leading to the activation of pathogen-specific B and T cell functions. In chronic HBV infection the anti-viral immune response fails to contain the virus and leads to persistent hepatic tissue damage which may finally result in liver cirrhosis and cancer. This T cell failure is associated with metabolic alterations suggesting that control of nutrient uptake and intracellular utilization as well as correct regulation of intracellular metabolic pathways are strategic for T cell differentiation during persistent chronic infections. This review will discuss some of the main features of the T cell metabolic processes which are relevant to the generation of an efficient antiviral response, with specific focus on their clinical relevance in chronic HBV infection in the perspective of possible strategies to correct deregulated metabolic pathways underlying T cell dysfunction of chronic HBV patients.
Collapse
Affiliation(s)
- Valeria Barili
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Carolina Boni
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Marzia Rossi
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Andrea Vecchi
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Alessandra Zecca
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Amalia Penna
- Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Gabriele Missale
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Carlo Ferrari
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy.
| | - Paola Fisicaro
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Laboratory of Viral Immunopathology, Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| |
Collapse
|
191
|
Kong Z, Zhou C, Kang J, Tan Z. Comparison of the Effects of Nonprotein and Protein Nitrogen on Apoptosis and Autophagy of Rumen Epithelial Cells in Goats. Animals (Basel) 2020; 10:E2079. [PMID: 33182520 PMCID: PMC7696569 DOI: 10.3390/ani10112079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/05/2020] [Indexed: 12/04/2022] Open
Abstract
Protein nutrition is particularly important for the self-renewal processes of gastrointestinal epithelial cells. The self-renewal of cells is inseparable from the interaction between apoptosis and autophagy. However, there are few reports on the relationship between different nitrogen sources and apoptosis/autophagy. In this study, the relative protein expression of Bcl-2-associated X protein(Bax), caspase-3, and p62 was significantly higher (p < 0.05), while that of Bcl-xl, Bcl-2, Beclin1, and Microtuble-associated protein light chain 3 (LC3-II) was significantly lower (p < 0.05), in the NH4Cl group in comparison with the NH4Cl + 4-phenylbutyric acid (4PBA) group. In addition, the relative protein expression of Bax and caspase-3 was significantly higher (p < 0.05), while that of Bcl-2 and Bcl-xl was decreased significantly (p < 0.05), in the NH4Cl + 3-Methyladenine (3-MA) group and the methionine (Met) + 3-MA group in comparison with the NH4Cl group. Furthermore, the relative protein expression of Beclin1 and LC3B-II was significantly lower (p < 0.05), while that of p62 was significantly higher (p < 0.05), in the NH4Cl + Z-VAD-FMK group and the Met + Z-VAD-FMK group in comparison with the NH4Cl group. In conclusion, our results suggested that endoplasmic reticulum (ER) stress played a critical role in the crosstalk between apoptosis and autophagy induced by NH4Cl and Met. Autophagy had a more obvious ameliorative effect on ruminal epithelial cell apoptosis after treatment with nonprotein nitrogen than after treatment with protein nitrogen. These findings may reveal the molecular mechanism of apoptosis and autophagy induced by nonprotein nitrogen and protein nitrogen.
Collapse
Affiliation(s)
- Zhiwei Kong
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Z.K.); (J.K.); (Z.T.)
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- College of Food Engineering and Biotechnology, Han Shan Normal University, Chaozhou 521041, China
| | - Chuanshe Zhou
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Z.K.); (J.K.); (Z.T.)
- Hunan Co-Innovation Center of Safety Animal Production, CICSAP, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410128, China
| | - Jinhe Kang
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Z.K.); (J.K.); (Z.T.)
- Hunan Co-Innovation Center of Safety Animal Production, CICSAP, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410128, China
| | - Zhiliang Tan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Z.K.); (J.K.); (Z.T.)
- Hunan Co-Innovation Center of Safety Animal Production, CICSAP, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410128, China
| |
Collapse
|
192
|
Zhao W, Ahmed S, Ahmed S, Yangliu Y, Wang H, Cai X. Analysis of long non-coding RNAs in epididymis of cattleyak associated with male infertility. Theriogenology 2020; 160:61-71. [PMID: 33181482 DOI: 10.1016/j.theriogenology.2020.10.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/07/2020] [Accepted: 10/26/2020] [Indexed: 02/08/2023]
Abstract
Cattleyak (CY), is a cross breed between cattle and yak (YK), which display equal adaptability to the harsh environment as YK and much higher performances than YK. However, the CY is female fertile and male sterile. Previous studies were conducted on testes tissues to investigate the mechanism of male infertility in CY. There is no systematic research on genes, especially lncRNAs between CY and YK epididymis. In this study, Illumina Hiseq was performed to profile the epididymis transcriptome (lncRNA and mRNA) of CY and YK. In total 18859 lncRNAs were identified, from which lincRNAs 12458, antisense lncRNAs 2345, intronic lncRNAs 3101, and sense lncRNAs 955 respectively. We have identified 345 DE lncRNAs and 3008 DE mRNAs between YK and CY epididymis. Thirteen DEGs were validated by quantitative real-time PCR. Combing with DEG, 14 couples of lncRNAs and their target genes were both DE, and 6 of them including CCDC39, KCNJ16, NECTIN2, MRPL20, PSMC4, and DEFB112 show their potential infertility-related terms such as cellular motility, sperm maturation, sperm storage, cellular junction, folate metabolism, and capacitation. On the other hand, several down-regulated genes such as DEFB124, DEFB126, DEFB125, DEFB127, DEFB129, CES5A, TKDP1, CST3, RNASE9 and CD52 in CY compared to YK were involved in the immune response and sperm maturation. Therefore, comprehensive analysis for lncRNAs and their target genes may enhance our understanding of the molecular mechanisms underlying the process of sperm maturation in CY and may provide important resources for further research.
Collapse
Affiliation(s)
- Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Saeed Ahmed
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Siraj Ahmed
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Yueling Yangliu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Hongmei Wang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization (Southwest Minzu University), Ministry of Education, Chengdu, Sichuan, 610041, China; Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
193
|
Mandarano AH, Maya J, Giloteaux L, Peterson DL, Maynard M, Gottschalk CG, Hanson MR. Myalgic encephalomyelitis/chronic fatigue syndrome patients exhibit altered T cell metabolism and cytokine associations. J Clin Invest 2020; 130:1491-1505. [PMID: 31830003 DOI: 10.1172/jci132185] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/26/2019] [Indexed: 12/25/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex disease with no known cause or mechanism. There is an increasing appreciation for the role of immune and metabolic dysfunction in the disease. ME/CFS has historically presented in outbreaks, often has a flu-like onset, and results in inflammatory symptoms. Patients suffer from severe fatigue and postexertional malaise. There is little known about the metabolism of specific immune cells in patients with ME/CFS. To investigate immune metabolism in ME/CFS, we isolated CD4+ and CD8+ T cells from 53 patients with ME/CFS and 45 healthy controls. We analyzed glycolysis and mitochondrial respiration in resting and activated T cells, along with markers related to cellular metabolism and plasma cytokines. We found that ME/CFS CD8+ T cells had reduced mitochondrial membrane potential compared with those from healthy controls. Both CD4+ and CD8+ T cells from patients with ME/CFS had reduced glycolysis at rest, whereas CD8+ T cells also had reduced glycolysis following activation. Patients with ME/CFS had significant correlations between measures of T cell metabolism and plasma cytokine abundance that differed from correlations seen in healthy control subjects. Our data indicate that patients have impaired T cell metabolism consistent with ongoing immune alterations in ME/CFS that may illuminate the mechanism behind this disease.
Collapse
Affiliation(s)
- Alexandra H Mandarano
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Jessica Maya
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Ludovic Giloteaux
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | | | - Marco Maynard
- Simmaron Research Institute, Incline Village, Nevada, USA
| | | | - Maureen R Hanson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| |
Collapse
|
194
|
Qu Y, Zhang S, Qu Y, Guo H, Wang S, Wang X, Huang T, Zhou H. Novel Gene Signature Reveals Prognostic Model in Acute Myeloid Leukemia. Front Genet 2020; 11:566024. [PMID: 33193652 PMCID: PMC7655922 DOI: 10.3389/fgene.2020.566024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/08/2020] [Indexed: 01/23/2023] Open
Abstract
Background Acute myeloid leukemia (AML) is a clonal malignant disease with poor prognosis and a low overall survival rate. Although many studies on the treatment and detection of AML have been conducted, the molecular mechanism of AML development and progression has not been fully elucidated. The present study was designed to pursuit the molecular mechanism of AML using a comprehensive bioinformatics analysis, and build an applicable model to predict the survival probability of AML patients in clinical use. Methods To simplify the complicated regulatory networks, we performed the gene co-expression and PPI network based on WGCNA and STRING database using modularization design. Two machine learning methods, A least absolute shrinkage and selector operation (LASSO) algorithm and support vector machine-recursive feature elimination (SVM-RFE), were used to filter the common hub genes by five-fold cross-validation. The candidate hub genes were used to build the predictive model of AML by the cox-proportional hazards analysis, and validated in The Cancer Genome Atlas (TCGA) cohort and ohsu cohort, which were reliable in the experimental verification by qRT-PCR and western blotting in mRNA and protein levels. Results Three hub genes, FLT3, CD177 and TTPAL were used to build a clinically applicable model to predict the survival probability of AML patients and divided them into high and low groups. To compare the survival ability of the model with the classical clinical features, we generated the nomogram. The model displayed the most risk points contrast to other clinical characteristics, which was compatible with the data of cox multivariate regression. Conclusion This study reveal the novel molecular mechanism of AML, and construct a clinical model significantly related to AML patient prognosis. We showed the integrated roles of critical pathways, hub genes associated, which provide potential targets and new research ideas for the treatment and early detection of AML.
Collapse
Affiliation(s)
- Ying Qu
- Department of Hematology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Shuying Zhang
- Department of Hematology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Yanzhang Qu
- Department of Hematology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Heng Guo
- Department of Hematology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Suling Wang
- Department of Hematology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Xuemei Wang
- Department of Hematology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Tianjiao Huang
- Department of Hematology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Hong Zhou
- Department of Hematology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| |
Collapse
|
195
|
Tan S, Li S, Min Y, Gisterå A, Moruzzi N, Zhang J, Sun Y, Andersson J, Malmström RE, Wang M, Berggren PO, Schlisio S, Liao W, Ketelhuth DFJ, Ma C, Li N. Platelet factor 4 enhances CD4 + T effector memory cell responses via Akt-PGC1α-TFAM signaling-mediated mitochondrial biogenesis. J Thromb Haemost 2020; 18:2685-2700. [PMID: 32671959 DOI: 10.1111/jth.15005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/10/2020] [Accepted: 07/08/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cell metabolism drives T cell functions, while platelets regulate overall CD4+ T cell immune responses. OBJECTIVE To investigate if platelets influence cell metabolism and thus regulate CD4+ T effector memory cell (Tem) responses. METHODS Human CD4+ Tem cells were activated with αCD3/αCD28 and cultured without or with platelets or platelet-derived mediators. RESULTS Polyclonal stimulation induced rapid and marked Th1 and Treg cell activation of CD4+ Tem cells. Platelet co-culture enhanced Th1 response transiently, while it persistently enhanced Treg cell activation of Tem cells, with an enhancement that plateaued by day 3. Platelet factor 4 (PF4) was the key platelet-derived mediator regulating CD4+ Tem cell responses, which involved cellular metabolisms as indicated by mass spectrometric analyses. PF4 exerted its effects via its receptor CXCR3, attenuated Akt activity, and reduced PGC1α phosphorylation, and resulted in elevations of PGC1α function and mitochondrial transcription factor A (TFAM) synthesis. The latter increased mitochondrial biogenesis, and subsequently enhanced Th1 and Treg responses. Consistent with these observations, inhibition of mitochondrial function by rotenone counteracted the enhancements by recombinant PF4, and TFAM overexpression by TFAM-adenovirus infection mimicked PF4 effects. Furthermore, increased mitochondrial mass elevated oxygen consumption, and enhanced adenosine triphosphate and reactive oxygen species production, which, in turn, stimulated Th1 (T-bet) and Treg (FoxP3) transcription factor expression and corresponding CD4+ T effector cell responses. CONCLUSIONS Platelets enhance CD4+ T cell responses of Tem cells through PF4-dependent and Akt-PGC1α-TFAM signaling-mediated mitochondrial biogenesis. Hence, PF4 may be a promising intervention target of platelet-regulated immune responses.
Collapse
Affiliation(s)
- Shuai Tan
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| | - Shuijie Li
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yanan Min
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| | - Anton Gisterå
- Department of Medicine-Solna, Cardiovascular Medicine Unit, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Noah Moruzzi
- Department of Molecular Medicine and Surgery, Rolf Ruft Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Junhao Zhang
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Sun
- Shandong University Cheeloo Medical College, Institute of Immunology, Jinan, China
| | - John Andersson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rickard E Malmström
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
- Department of Laboratory Medicine, Clinical Pharmacology, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Miao Wang
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Per-Olof Berggren
- Department of Molecular Medicine and Surgery, Rolf Ruft Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Schlisio
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Daniel F J Ketelhuth
- Department of Medicine-Solna, Cardiovascular Medicine Unit, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Chunhong Ma
- Shandong University Cheeloo Medical College, Institute of Immunology, Jinan, China
| | - Nailin Li
- Department of Medicine-Solna, Clinical Epidemiology Unit, Clinical Pharmacology Group, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
196
|
De Lira MN, Raman SJ, Schulze A, Schneider-Schaulies S, Avota E. Neutral Sphingomyelinase-2 (NSM 2) Controls T Cell Metabolic Homeostasis and Reprogramming During Activation. Front Mol Biosci 2020; 7:217. [PMID: 33088808 PMCID: PMC7498697 DOI: 10.3389/fmolb.2020.00217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
Neutral sphingomyelinase-2 (NSM2) is a member of a superfamily of enzymes responsible for conversion of sphingomyelin into phosphocholine and ceramide at the cytosolic leaflet of the plasma membrane. Upon specific ablation of NSM2, T cells proved to be hyper-responsive to CD3/CD28 co-stimulation, indicating that the enzyme acts to dampen early overshooting activation of these cells. It remained unclear whether hyper-reactivity of NSM2-deficient T cells is supported by a deregulated metabolic activity in these cells. Here, we demonstrate that ablation of NSM2 activity affects metabolism of the quiescent CD4+ T cells which accumulate ATP in mitochondria and increase basal glycolytic activity. This supports enhanced production of total ATP and metabolic switch early after TCR/CD28 stimulation. Most interestingly, increased metabolic activity in resting NSM2-deficient T cells does not support sustained response upon stimulation. While elevated under steady-state conditions in NSM2-deficient CD4+ T cells, the mTORC1 pathway regulating mitochondria size, oxidative phosphorylation, and ATP production is impaired after 24 h of stimulation. Taken together, the absence of NSM2 promotes a hyperactive metabolic state in unstimulated CD4+ T cells yet fails to support sustained T cell responses upon antigenic stimulation.
Collapse
Affiliation(s)
| | | | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg, Germany
| | | | - Elita Avota
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
197
|
SIRT5 Contributes to Colorectal Cancer Growth by Regulating T Cell Activity. J Immunol Res 2020; 2020:3792409. [PMID: 32953892 PMCID: PMC7481950 DOI: 10.1155/2020/3792409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022] Open
Abstract
Over the past several years, SIRT5 has attracted considerable attention in metabolic regulation. However, the function of SIRT5 in tumorigenesis by regulating tumor microenvironment is poorly understood. In this work, we found that Sirt5 knockout mice were resistant to AOM and DSS-induced colitis-associated colorectal tumorigenesis and the level of IFN-γ in their tumor microenvironment was higher. Additionally, proteome and network analysis revealed that SIRT5 was important in the T cell receptor signaling pathway. Furthermore, we determined that a deficiency of Sirt5 induced stronger T cell activation and demonstrated that SIRT5 played a pivotal role in regulating the differentiation of CD4+ regulatory T (Treg) cells and T helper 1 (Th1) cells. An imbalance in the lineages of immunosuppressive Treg cells and the inflammatory Th1 subsets of helper T cells leads to the development of colon cancer. Our results revealed a regulatory role of SIRT5 in T cell activation and colorectal tumorigenesis.
Collapse
|
198
|
Hao Shi, Yan KK, Ding L, Qian C, Chi H, Yu J. Network Approaches for Dissecting the Immune System. iScience 2020; 23:101354. [PMID: 32717640 PMCID: PMC7390880 DOI: 10.1016/j.isci.2020.101354] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/21/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
The immune system is a complex biological network composed of hierarchically organized genes, proteins, and cellular components that combat external pathogens and monitor the onset of internal disease. To meet and ultimately defeat these challenges, the immune system orchestrates an exquisitely complex interplay of numerous cells, often with highly specialized functions, in a tissue-specific manner. One of the major methodologies of systems immunology is to measure quantitatively the components and interaction levels in the immunologic networks to construct a computational network and predict the response of the components to perturbations. The recent advances in high-throughput sequencing techniques have provided us with a powerful approach to dissecting the complexity of the immune system. Here we summarize the latest progress in integrating omics data and network approaches to construct networks and to infer the underlying signaling and transcriptional landscape, as well as cell-cell communication, in the immune system, with a focus on hematopoiesis, adaptive immunity, and tumor immunology. Understanding the network regulation of immune cells has provided new insights into immune homeostasis and disease, with important therapeutic implications for inflammation, cancer, and other immune-mediated disorders.
Collapse
Affiliation(s)
- Hao Shi
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Koon-Kiu Yan
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Liang Ding
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Chenxi Qian
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jiyang Yu
- Departments of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
199
|
Vinogradova EV, Zhang X, Remillard D, Lazar DC, Suciu RM, Wang Y, Bianco G, Yamashita Y, Crowley VM, Schafroth MA, Yokoyama M, Konrad DB, Lum KM, Simon GM, Kemper EK, Lazear MR, Yin S, Blewett MM, Dix MM, Nguyen N, Shokhirev MN, Chin EN, Lairson LL, Melillo B, Schreiber SL, Forli S, Teijaro JR, Cravatt BF. An Activity-Guided Map of Electrophile-Cysteine Interactions in Primary Human T Cells. Cell 2020; 182:1009-1026.e29. [PMID: 32730809 PMCID: PMC7775622 DOI: 10.1016/j.cell.2020.07.001] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/14/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022]
Abstract
Electrophilic compounds originating from nature or chemical synthesis have profound effects on immune cells. These compounds are thought to act by cysteine modification to alter the functions of immune-relevant proteins; however, our understanding of electrophile-sensitive cysteines in the human immune proteome remains limited. Here, we present a global map of cysteines in primary human T cells that are susceptible to covalent modification by electrophilic small molecules. More than 3,000 covalently liganded cysteines were found on functionally and structurally diverse proteins, including many that play fundamental roles in immunology. We further show that electrophilic compounds can impair T cell activation by distinct mechanisms involving the direct functional perturbation and/or degradation of proteins. Our findings reveal a rich content of ligandable cysteines in human T cells and point to electrophilic small molecules as a fertile source for chemical probes and ultimately therapeutics that modulate immunological processes and their associated disorders.
Collapse
Affiliation(s)
| | - Xiaoyu Zhang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David Remillard
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel C Lazar
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Radu M Suciu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yujia Wang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Giulia Bianco
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yu Yamashita
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; Medicinal Chemistry Research Laboratories, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd., 463-10 Kawauchi-cho, Tokushima 771-0192, Japan
| | - Vincent M Crowley
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael A Schafroth
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Minoru Yokoyama
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David B Konrad
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kenneth M Lum
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gabriel M Simon
- Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA 92121, USA
| | - Esther K Kemper
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael R Lazear
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sifei Yin
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Megan M Blewett
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Melissa M Dix
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nhan Nguyen
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Emily N Chin
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bruno Melillo
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02138, USA
| | - Stuart L Schreiber
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John R Teijaro
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
200
|
Wang Z, Kavdia K, Dey KK, Pagala VR, Kodali K, Liu D, Lee DG, Sun H, Chepyala SR, Cho JH, Niu M, High AA, Peng J. High-throughput and Deep-proteome Profiling by 16-plex Tandem Mass Tag Labeling Coupled with Two-dimensional Chromatography and Mass Spectrometry. J Vis Exp 2020:10.3791/61684. [PMID: 32894271 PMCID: PMC7752892 DOI: 10.3791/61684] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Isobaric tandem mass tag (TMT) labeling is widely used in proteomics because of its high multiplexing capacity and deep proteome coverage. Recently, an expanded 16-plex TMT method has been introduced, which further increases the throughput of proteomic studies. In this manuscript, we present an optimized protocol for 16-plex TMT-based deep-proteome profiling, including protein sample preparation, enzymatic digestion, TMT labeling reaction, two-dimensional reverse-phase liquid chromatography (LC/LC) fractionation, tandem mass spectrometry (MS/MS), and computational data processing. The crucial quality control steps and improvements in the process specific for the 16-plex TMT analysis are highlighted. This multiplexed process offers a powerful tool for profiling a variety of complex samples such as cells, tissues, and clinical specimens. More than 10,000 proteins and posttranslational modifications such as phosphorylation, methylation, acetylation, and ubiquitination in highly complex biological samples from up to 16 different samples can be quantified in a single experiment, providing a potent tool for basic and clinical research.
Collapse
Affiliation(s)
- Zhen Wang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital
| | - Kanisha Kavdia
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital
| | - Kaushik Kumar Dey
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital
| | | | - Kiran Kodali
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital
| | - Danting Liu
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital
| | - Dong Geun Lee
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital
| | - Huan Sun
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital
| | - Surendhar Reddy Chepyala
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital
| | - Ji-Hoon Cho
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital
| | - Mingming Niu
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital
| | - Anthony A High
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital;
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital; Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital;
| |
Collapse
|