1
|
Balestra F, De Luca M, Panzetta G, Depalo N, Rizzi F, Mastrogiacomo R, Coletta S, Serino G, Piccinno E, Stabile D, Pesole PL, De Nunzio V, Pinto G, Cerabino N, Di Chito M, Notarnicola M, Shahini E, De Pergola G, Scavo MP. An 8-Week Very Low-Calorie Ketogenic Diet (VLCKD) Alters the Landscape of Obese-Derived Small Extracellular Vesicles (sEVs), Redefining Hepatic Cell Phenotypes. Nutrients 2024; 16:4189. [PMID: 39683581 DOI: 10.3390/nu16234189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Background. Very low-calorie ketogenic diets (VLCKD) are an effective weight-loss strategy for obese individuals, reducing risks of liver conditions such as non-alcoholic steatohepatitis and fibrosis. Small extracellular vesicles (sEVs) are implicated in liver fibrosis by influencing hepatic cell phenotypes and contributing to liver damage. This study investigates sEVs derived from serum of 60 obese adults categorized into low fibrosis risk (LR) and intermediate/high fibrosis risk (IHR) groups based on FibroScan elastography (FIB E scores, limit value 8 kPa) and all participants underwent an 8-week VLCKD intervention. Methods. The study examines the impact of these sEVs on fibrosis markers, inflammation, and autophagy in a hepatocyte cell line (HEPA-RG) using bioinformatics, RNA sequencing, lipidomics, RT-PCR, and Western blotting before (T0) and after (T1) VLCKD. Results. sEVs from LR patients post-VLCKD reduced fibrosis related gene expression (e.g., ACTA2) and enhanced proteins associated with regeneration and inflammation (e.g., HDAC6). Conversely, sEVs from IHR patients increased fibrosis and inflammation related gene expression (PIK3CB, AKT1, ACTA2) in hepatocytes, raising concerns about VLCKD suitability for IHR patients. IHR sEVs also decreased expression of HDAC10, HDAC6, HDAC3, MMP19, and MMP2, while increasing modulation of p-AKT, α-SMA, and VIM. Conclusion. These findings underscore the critical role of sEVs in regulating inflammation, remodeling, and hepatic stress responses, particularly in IHR patients, and suggest sEVs could complement instrumental evaluations like FibroScan in fibrosis assessment.
Collapse
Affiliation(s)
- Francesco Balestra
- Laboratory of Molecular Medicine, National Institute of Gastroenterology IRCCS "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Maria De Luca
- Laboratory of Molecular Medicine, National Institute of Gastroenterology IRCCS "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Giorgia Panzetta
- Laboratory of Molecular Medicine, National Institute of Gastroenterology IRCCS "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Nicoletta Depalo
- Institute for Chemical-Physical Processes, Italian National Research Council (IPCF)-CNR SS Bari, Via Orabona 4, 70125 Bari, Italy
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Bari Research Unit, Via Orabona 4, 70126 Bari, Italy
| | - Federica Rizzi
- Institute for Chemical-Physical Processes, Italian National Research Council (IPCF)-CNR SS Bari, Via Orabona 4, 70125 Bari, Italy
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Bari Research Unit, Via Orabona 4, 70126 Bari, Italy
| | - Rita Mastrogiacomo
- Department of Chemistry, University of Bari, Via Orabona 4, 70125 Bari, Italy
| | - Sergio Coletta
- Core Facility Biobank, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Grazia Serino
- Laboratory of Molecular Medicine, National Institute of Gastroenterology IRCCS "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Emanuele Piccinno
- Laboratory of Molecular Medicine, National Institute of Gastroenterology IRCCS "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Dolores Stabile
- Core Facility Biobank, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Pasqua Letizia Pesole
- Core Facility Biobank, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Valentina De Nunzio
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology, "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Giuliano Pinto
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology, "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Nicole Cerabino
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS "Saverio de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Martina Di Chito
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS "Saverio de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Maria Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology, "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Giovanni De Pergola
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS "Saverio de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Maria Principia Scavo
- Laboratory of Molecular Medicine, National Institute of Gastroenterology IRCCS "S. de Bellis", Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| |
Collapse
|
2
|
Gómez-Morón Á, Tsukalov I, Scagnetti C, Pertusa C, Lozano-Prieto M, Martínez-Fleta P, Requena S, Martín P, Alfranca A, Martin-Gayo E, Martin-Cofreces NB. Cytosolic protein translation regulates cell asymmetry and function in early TCR activation of human CD8 + T lymphocytes. Front Immunol 2024; 15:1411957. [PMID: 39114656 PMCID: PMC11303187 DOI: 10.3389/fimmu.2024.1411957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction CD8+ cytotoxic T lymphocytes (CTLs) are highly effective in defending against viral infections and tumours. They are activated through the recognition of peptide-MHC-I complex by the T-cell receptor (TCR) and co-stimulation. This cognate interaction promotes the organisation of intimate cell-cell connections that involve cytoskeleton rearrangement to enable effector function and clearance of the target cell. This is key for the asymmetric transport and mobilisation of lytic granules to the cell-cell contact, promoting directed secretion of lytic mediators such as granzymes and perforin. Mitochondria play a role in regulating CTL function by controlling processes such as calcium flux, providing the necessary energy through oxidative phosphorylation, and its own protein translation on 70S ribosomes. However, the effect of acute inhibition of cytosolic translation in the rapid response after TCR has not been studied in mature CTLs. Methods Here, we investigated the importance of cytosolic protein synthesis in human CTLs after early TCR activation and CD28 co-stimulation for the dynamic reorganisation of the cytoskeleton, mitochondria, and lytic granules through short-term chemical inhibition of 80S ribosomes by cycloheximide and 80S and 70S by puromycin. Results We observed that eukaryotic ribosome function is required to allow proper asymmetric reorganisation of the tubulin cytoskeleton and mitochondria and mTOR pathway activation early upon TCR activation in human primary CTLs. Discussion Cytosolic protein translation is required to increase glucose metabolism and degranulation capacity upon TCR activation and thus to regulate the full effector function of human CTLs.
Collapse
Affiliation(s)
- Álvaro Gómez-Morón
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Ilya Tsukalov
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Medicine Faculty, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Camila Scagnetti
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Videomicroscopy Unit, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Clara Pertusa
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Marta Lozano-Prieto
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Pedro Martínez-Fleta
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Silvia Requena
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Pilar Martín
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Area of Vascular Pathophysiology, Laboratory of Regulatory Molecules of Inflammatory Processes, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, Spain
| | - Aranzazu Alfranca
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Medicine Faculty, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Martin-Gayo
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Medicine Faculty, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFECC), Instituto de Salud Carlos III, Madrid, Spain
| | - Noa B Martin-Cofreces
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS- Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Videomicroscopy Unit, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, Spain
| |
Collapse
|
3
|
Ducellier S, Demeules M, Letribot B, Gaetani M, Michaudel C, Sokol H, Hamze A, Alami M, Nascimento M, Apcher S. Dual molecule targeting HDAC6 leads to intratumoral CD4+ cytotoxic lymphocytes recruitment through MHC-II upregulation on lung cancer cells. J Immunother Cancer 2024; 12:e007588. [PMID: 38609101 PMCID: PMC11015306 DOI: 10.1136/jitc-2023-007588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Despite the current therapeutic treatments including surgery, chemotherapy, radiotherapy and more recently immunotherapy, the mortality rate of lung cancer stays high. Regarding lung cancer, epigenetic modifications altering cell cycle, angiogenesis and programmed cancer cell death are therapeutic targets to combine with immunotherapy to improve treatment success. In a recent study, we uncovered that a molecule called QAPHA ((E)-3-(5-((2-cyanoquinolin-4-yl)(methyl)amino)-2-methoxyphenyl)-N-hydroxyacrylamide) has a dual function as both a tubulin polymerization and HDAC inhibitors. Here, we investigate the impact of this novel dual inhibitor on the immune response to lung cancer. METHODS To elucidate the mechanism of action of QAPHA, we conducted a chemical proteomics analysis. Using an in vivo mouse model of lung cancer (TC-1 tumor cells), we assessed the effects of QAPHA on tumor regression. Tumor infiltrating immune cells were characterized by flow cytometry. RESULTS In this study, we first showed that QAPHA effectively inhibited histone deacetylase 6, leading to upregulation of HSP90, cytochrome C and caspases, as revealed by proteomic analysis. We confirmed that QAPHA induces immunogenic cell death (ICD) by expressing calreticulin at cell surface in vitro and demonstrated its efficacy as a vaccine in vivo. Remarkably, even at a low concentration (0.5 mg/kg), QAPHA achieved complete tumor regression in approximately 60% of mice treated intratumorally, establishing a long-lasting anticancer immune response. Additionally, QAPHA treatment promoted the infiltration of M1-polarized macrophages in treated mice, indicating the induction of a pro-inflammatory environment within the tumor. Very interestingly, our findings also revealed that QAPHA upregulated major histocompatibility complex class II (MHC-II) expression on TC-1 tumor cells both in vitro and in vivo, facilitating the recruitment of cytotoxic CD4+T cells (CD4+CTL) expressing CD4+, NKG2D+, CRTAM+, and Perforin+. Finally, we showed that tumor regression strongly correlates to MHC-II expression level on tumor cell and CD4+ CTL infiltrate. CONCLUSION Collectively, our findings shed light on the discovery of a new multitarget inhibitor able to induce ICD and MHC-II upregulation in TC-1 tumor cell. These two processes participate in enhancing a specific CD4+ cytotoxic T cell-mediated antitumor response in vivo in our model of lung cancer. This breakthrough suggests the potential of QAPHA as a promising agent for cancer treatment.
Collapse
Affiliation(s)
- Sarah Ducellier
- UMR 1015 Immunologie des tumeurs et immunothérapie contre le cancer, B2M, Gustave Roussy, Villejuif, France
| | - Mélanie Demeules
- UMR 1015 Immunologie des tumeurs et immunothérapie contre le cancer, B2M, Gustave Roussy, Villejuif, France
| | | | - Massimiliano Gaetani
- Chemical Proteomics Core Facility, Division of Chemistry I Department of Medical Biochemistry andBiophysics, Karolinska Institute, Stockholm, Sweden
- Chemical Proteomics Unit, Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
- Chemical Proteomics, Swedish National Infrastructure for Biological Mass Spectrometry (BioMS), Stockholm, Sweden
| | - Chloé Michaudel
- AgroParisTech Micalis institute, INRAe Université Paris-Saclay, Jouy-en-Josas, France
| | - Harry Sokol
- Gastroenterology Department, Centre de Recherche Saint-Antoine Sorbonne Université, INSERM CRSA, AP-HP, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | | | - Mouad Alami
- BioCIS, CNRS Université Paris-Saclay, Orsay, France
| | - Mégane Nascimento
- UMR 1015 Immunologie des tumeurs et immunothérapie contre le cancer, B2M, Gustave Roussy, Villejuif, France
| | - Sébastien Apcher
- UMR 1015 Immunologie des tumeurs et immunothérapie contre le cancer, B2M, Gustave Roussy, Villejuif, France
| |
Collapse
|
4
|
Lee JY, Ma HW, Kim JH, Park IS, Son M, Ryu KH, Shin J, Kim SW, Cheon JH. Novel Histone Deacetylase 6 Inhibitor Confers Anti-inflammatory Effects and Enhances Gut Barrier Function. Gut Liver 2023; 17:766-776. [PMID: 36167345 PMCID: PMC10502503 DOI: 10.5009/gnl220159] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/07/2022] [Accepted: 07/18/2022] [Indexed: 11/04/2022] Open
Abstract
Background/Aims The purpose of the current study was to examine the anti-inflammatory effects of CKD-506, a novel histone deacetylase 6 inhibitor, on human peripheral blood mononuclear cells (PBMCs) and CD4+ T cells and to explore the relationship between CKD-506 and gut epithelial barrier function. Methods Lipopolysaccharide-stimulated human PBMCs from inflammatory bowel disease (IBD) patients were treated with CKD-506, and tumor necrosis factor (TNF)-α expression was measured using an enzyme-linked immunosorbent assay. The proliferation of CD4+ T cells from IBD patients was evaluated using flow cytometric analysis. The effects of CKD-506 on gut barrier function in a cell line and colon organoids, based on examinations of mRNA production, goblet cell differentiation, and E-cadherin recovery, were investigated using quantitative reverse transcription polymerase chain reaction, immunofluorescence, and a fluorescein isothiocyanate-dextran permeability assay. Results Secretion of TNF-α, a pivotal pro-inflammatory mediator in IBD, by lipopolysaccharide-triggered PBMCs was markedly decreased by CKD-506 treatment in a dose-dependent manner and to a greater extent than by tofacitinib or tubastatin A treatment. E-cadherin mRNA expression and goblet cell differentiation increased significantly and dose-dependently in HT-29 cells in response to CKD-506, and inhibition of E-cadherin loss after TNF-α stimulation was significantly reduced both in HT-29 cells and gut organoids. Caco-2 cells treated with CKD-506 showed a significant reduction in barrier permeability in a dose-dependent manner. Conclusions The present study demonstrated that CKD-506 has anti-inflammatory effects on PBMCs and CD4 T cells and improves gut barrier function, suggesting its potential as a small-molecule therapeutic option for IBD.
Collapse
Affiliation(s)
- Jae-Young Lee
- Department of Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Woo Ma
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyung Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Mijeong Son
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Keun Ho Ryu
- Department of Non-Clinical Study, CKD Research Institute, CKD Pharmaceutical Co., Yongin, Korea
| | - Jieun Shin
- Department of Non-Clinical Study, CKD Research Institute, CKD Pharmaceutical Co., Yongin, Korea
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
5
|
Zhang QQ, Zhang WJ, Chang S. HDAC6 inhibition: a significant potential regulator and therapeutic option to translate into clinical practice in renal transplantation. Front Immunol 2023; 14:1168848. [PMID: 37545520 PMCID: PMC10401441 DOI: 10.3389/fimmu.2023.1168848] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 05/30/2023] [Indexed: 08/08/2023] Open
Abstract
Histone deacetylase 6 (HDAC6), an almost exclusively cytoplasmic enzyme, plays an essential role in many biological processes and exerts its deacetylation-dependent/independent effects on a variety of target molecules, which has contributed to the flourishing growth of relatively isoform-specific enzyme inhibitors. Renal transplantation (RT) is one of the alternatively preferred treatments and the most cost-effective treatment approaches for the great majority of patients with end-stage renal disease (ESRD). HDAC6 expression and activity have recently been shown to be increased in kidney disease in a number of studies. To date, a substantial amount of validated studies has identified HDAC6 as a pivotal modulator of innate and adaptive immunity, and HDAC6 inhibitors (HDAC6i) are being developed and investigated for use in arrays of immune-related diseases, making HDAC6i a promising therapeutic candidate for the management of a variety of renal diseases. Based on accumulating evidence, HDAC6i markedly open up new avenues for therapeutic intervention to protect against oxidative stress-induced damage, tip the balance in favor of the generation of tolerance-related immune cells, and attenuate fibrosis by inhibiting multiple activations of cell profibrotic signaling pathways. Taken together, we have a point of view that targeting HDAC6 may be a novel approach for the therapeutic strategy of RT-related complications, including consequences of ischemia-reperfusion injury, induction of immune tolerance in transplantation, equilibrium of rejection, and improvement of chronic renal graft interstitial fibrosis after transplantation in patients. Herein, we will elaborate on the unique function of HDAC6, which focuses on therapeutical mechanism of action related to immunological events with a general account of the tantalizing potential to the clinic.
Collapse
Affiliation(s)
- Qian-qian Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Wei-jie Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Sheng Chang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
6
|
Izquierdo-Serrano R, Fernández-Delgado I, Moreno-Gonzalo O, Martín-Gayo E, Calzada-Fraile D, Ramírez-Huesca M, Jorge I, Camafeita E, Abián J, Vicente-Manzanares M, Veiga E, Vázquez J, Sánchez-Madrid F. Extracellular vesicles from Listeria monocytogenes-infected dendritic cells alert the innate immune response. Front Immunol 2022; 13:946358. [PMID: 36131943 PMCID: PMC9483171 DOI: 10.3389/fimmu.2022.946358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Communication through cell-cell contacts and extracellular vesicles (EVs) enables immune cells to coordinate their responses against diverse types of pathogens. The function exerted by EVs in this context depends on the proteins and nucleic acids loaded into EVs, which elicit specific responses involved in the resolution of infection. Several mechanisms control protein and nucleic acid loading into EVs; in this regard, acetylation has been described as a mechanism of cellular retention during protein sorting to exosomes. HDAC6 is a deacetylase involved in the control of cytoskeleton trafficking, organelle polarity and cell migration, defense against Listeria monocytogenes (Lm) infection and other immune related functions. Here, we show that the protein content of dendritic cells (DCs) and their secreted EVs (DEVs) vary during Lm infection, is enriched in proteins related to antiviral functions compared to non-infected cells and depends on HDAC6 expression. Analyses of the post-translational modifications revealed an alteration of the acetylation and ubiquitination profiles upon Lm infection both in DC lysates and DEVs. Functionally, EVs derived from infected DCs upregulate anti-pathogenic genes (e.g. inflammatory cytokines) in recipient immature DCs, which translated into protection from subsequent infection with vaccinia virus. Interestingly, absence of Listeriolysin O in Lm prevents DEVs from inducing this anti-viral state. In summary, these data underscore a new mechanism of communication between bacteria-infected DC during infection as they alert neighboring, uninfected DCs to promote antiviral responses.
Collapse
Affiliation(s)
- Raúl Izquierdo-Serrano
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
| | - Irene Fernández-Delgado
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Department of Immunology, Instituto Investigación Sanitaria Hospital Universitario La Princesa (IIS-HUP), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Olga Moreno-Gonzalo
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Department of Immunology, Instituto Investigación Sanitaria Hospital Universitario La Princesa (IIS-HUP), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Enrique Martín-Gayo
- Department of Immunology, Instituto Investigación Sanitaria Hospital Universitario La Princesa (IIS-HUP), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Diego Calzada-Fraile
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
| | - Marta Ramírez-Huesca
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Department of Immunology, Instituto Investigación Sanitaria Hospital Universitario La Princesa (IIS-HUP), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Inmaculada Jorge
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Emilio Camafeita
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Joaquín Abián
- Biological and Environmental Proteomics, Institut d’Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Salamanca, Spain
| | - Esteban Veiga
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Jesús Vázquez
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Department of Immunology, Instituto Investigación Sanitaria Hospital Universitario La Princesa (IIS-HUP), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- *Correspondence: Francisco Sánchez-Madrid,
| |
Collapse
|
7
|
Ham H, Medlyn M, Billadeau DD. Locked and Loaded: Mechanisms Regulating Natural Killer Cell Lytic Granule Biogenesis and Release. Front Immunol 2022; 13:871106. [PMID: 35558071 PMCID: PMC9088006 DOI: 10.3389/fimmu.2022.871106] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022] Open
Abstract
NK cell-mediated cytotoxicity is a critical element of our immune system required for protection from microbial infections and cancer. NK cells bind to and eliminate infected or cancerous cells via direct secretion of cytotoxic molecules toward the bound target cells. In this review, we summarize the current understanding of the molecular regulations of NK cell cytotoxicity, focusing on lytic granule development and degranulation processes. NK cells synthesize apoptosis-inducing proteins and package them into specialized organelles known as lytic granules (LGs). Upon activation of NK cells, LGs converge with the microtubule organizing center through dynein-dependent movement along microtubules, ultimately polarizing to the cytotoxic synapse where they subsequently fuse with the NK plasma membrane. From LGs biogenesis to degranulation, NK cells utilize several strategies to protect themselves from their own cytotoxic molecules. Additionally, molecular pathways that enable NK cells to perform serial killing are beginning to be elucidated. These advances in the understanding of the molecular pathways behind NK cell cytotoxicity will be important to not only improve current NK cell-based anti-cancer therapies but also to support the discovery of additional therapeutic opportunities.
Collapse
Affiliation(s)
- Hyoungjun Ham
- Division of Oncology Research, Mayo Clinic, Rochester, MN, United States
| | - Michael Medlyn
- Department of Immunology College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Daniel D Billadeau
- Division of Oncology Research, Mayo Clinic, Rochester, MN, United States.,Department of Immunology College of Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
8
|
Nanamori H, Sawada Y. Epigenetic Modification of PD-1/PD-L1-Mediated Cancer Immunotherapy against Melanoma. Int J Mol Sci 2022; 23:ijms23031119. [PMID: 35163049 PMCID: PMC8835029 DOI: 10.3390/ijms23031119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Malignant melanoma is one of the representative skin cancers with unfavorable clinical behavior. Immunotherapy is currently used for the treatment, and it dramatically improves clinical outcomes in patients with advanced malignant melanoma. On the other hand, not all these patients can obtain therapeutic efficacy. To overcome this limitation of current immunotherapy, epigenetic modification is a highlighted issue for clinicians. Epigenetic modification is involved in various physiological and pathological conditions in the skin. Recent studies identified that skin cancer, especially malignant melanoma, has advantages in tumor development, indicating that epigenetic manipulation for regulation of gene expression in the tumor can be expected to result in additional therapeutic efficacy during immunotherapy. In this review, we focus on the detailed molecular mechanism of epigenetic modification in immunotherapy, especially anti-PD-1/PD-L1 antibody treatment for malignant melanoma.
Collapse
|
9
|
Montacchiesi G, Pace L. Epigenetics and CD8 + T cell memory. Immunol Rev 2021; 305:77-89. [PMID: 34923638 DOI: 10.1111/imr.13057] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022]
Abstract
Following antigen recognition, CD8+ T lymphocytes can follow different patterns of differentiation, with the generation of different subsets characterized by distinct phenotypes, functions, and migration properties. The changes of transcription factors activity and chromatin structure dynamics drive the functional differentiation and phenotypic heterogeneity of these T cell subsets, which include short-lived effectors, long-term survival of memory, and also dysfunctional exhausted T cells. Recent progress in the field has shed light on the key contribution of chromatin organization to control the T cell fate specification. In fact, the understanding of these processes has important implications for the development of new immunotherapy protocols and to design new vaccination strategies. Here, we review the current understanding of the contribution of chromatin architecture and transcription factor activity orchestrating the gene expression programs guiding the CD8+ T cell subset commitment. We will focus on epigenetic changes, acting sequentially or in combination, which control the transcriptional programs governing T cell plasticity, stability, and memory. New molecular insights into the mechanisms of maintenance of cellular memory and identity, favoring or impeding the reprogramming, will be discussed in the context of T cell memory differentiation in infection and cancer.
Collapse
Affiliation(s)
- Gaia Montacchiesi
- Armenise-Harvard Immune Regulation Unit, Italian Institute for Genomic Medicine, Turin, Italy.,Candiolo Cancer Institute, FPO-IRCCS Candiolo (Turin), Turin, Italy.,University of Turin, Turin, Italy
| | - Luigia Pace
- Armenise-Harvard Immune Regulation Unit, Italian Institute for Genomic Medicine, Turin, Italy.,University of Turin, Turin, Italy
| |
Collapse
|
10
|
Post-translational modifications and stabilization of microtubules regulate transport of viral factors during infections. Biochem Soc Trans 2021; 49:1735-1748. [PMID: 34436545 DOI: 10.1042/bst20210017] [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: 05/24/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/17/2022]
Abstract
Tubulin post-translational modifications (PTMs) constitute a source of diversity for microtubule (MT) functions, in addition to the different isotypes of α and β-tubulin acting as building blocks of MTs. Also, MT-associated proteins (MAPs) confer different characteristics to MTs. The combination of all these factors regulates the stability of these structures that act as rails to transport organelles within the cell, facilitating the association of motor complexes. All these functions are involved in crucial cellular processes in most cell types, ranging from spindle formation in mitosis to the defense against incoming cellular threats during phagocytosis mediated by immune cells. The regulation of MT dynamics through tubulin PTMs has evolved to depend on many different factors that act in a complex orchestrated manner. These tightly regulated processes are particularly relevant during the induction of effective immune responses against pathogens. Viruses have proved not only to hijack MTs and MAPs in order to favor an efficient infection, but also to induce certain PTMs that improve their cellular spread and lead to secondary consequences of viral processes. In this review, we offer a perspective on relevant MT-related elements exploited by viruses.
Collapse
|
11
|
Martín-Cófreces NB, Valpuesta JM, Sánchez-Madrid F. Folding for the Immune Synapse: CCT Chaperonin and the Cytoskeleton. Front Cell Dev Biol 2021; 9:658460. [PMID: 33912568 PMCID: PMC8075050 DOI: 10.3389/fcell.2021.658460] [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: 01/25/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Abstract
Lymphocytes rearrange their shape, membrane receptors and organelles during cognate contacts with antigen-presenting cells (APCs). Activation of T cells by APCs through pMHC-TCR/CD3 interaction (peptide-major histocompatibility complex-T cell receptor/CD3 complexes) involves different steps that lead to the reorganization of the cytoskeleton and organelles and, eventually, activation of nuclear factors allowing transcription and ultimately, replication and cell division. Both the positioning of the lymphocyte centrosome in close proximity to the APC and the nucleation of a dense microtubule network beneath the plasma membrane from the centrosome support the T cell's intracellular polarity. Signaling from the TCR is facilitated by this traffic, which constitutes an important pathway for regulation of T cell activation. The coordinated enrichment upon T cell stimulation of the chaperonin CCT (chaperonin-containing tailless complex polypeptide 1; also termed TRiC) and tubulins at the centrosome area support polarized tubulin polymerization and T cell activation. The proteasome is also enriched in the centrosome of activated T cells, providing a mechanism to balance local protein synthesis and degradation. CCT assists the folding of proteins coming from de novo synthesis, therefore favoring mRNA translation. The functional role of this chaperonin in regulating cytoskeletal composition and dynamics at the immune synapse is discussed.
Collapse
Affiliation(s)
- Noa Beatriz Martín-Cófreces
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autonoma Madrid (UAM), Instituto Investigacion Sanitaria-Instituto Princesa (IIS-IP), Madrid, Spain.,Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | - Francisco Sánchez-Madrid
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autonoma Madrid (UAM), Instituto Investigacion Sanitaria-Instituto Princesa (IIS-IP), Madrid, Spain.,Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
12
|
Kopf A, Kiermaier E. Dynamic Microtubule Arrays in Leukocytes and Their Role in Cell Migration and Immune Synapse Formation. Front Cell Dev Biol 2021; 9:635511. [PMID: 33634136 PMCID: PMC7900162 DOI: 10.3389/fcell.2021.635511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/18/2021] [Indexed: 01/13/2023] Open
Abstract
The organization of microtubule arrays in immune cells is critically important for a properly operating immune system. Leukocytes are white blood cells of hematopoietic origin, which exert effector functions of innate and adaptive immune responses. During these processes the microtubule cytoskeleton plays a crucial role for establishing cell polarization and directed migration, targeted secretion of vesicles for T cell activation and cellular cytotoxicity as well as the maintenance of cell integrity. Considering this large spectrum of distinct effector functions, leukocytes require flexible microtubule arrays, which timely and spatially reorganize allowing the cells to accommodate their specific tasks. In contrast to other specialized cell types, which typically nucleate microtubule filaments from non-centrosomal microtubule organizing centers (MTOCs), leukocytes mainly utilize centrosomes for sites of microtubule nucleation. Yet, MTOC localization as well as microtubule organization and dynamics are highly plastic in leukocytes thus allowing the cells to adapt to different environmental constraints. Here we summarize our current knowledge on microtubule organization and dynamics during immune processes and how these microtubule arrays affect immune cell effector functions. We particularly highlight emerging concepts of microtubule involvement during maintenance of cell shape and physical coherence.
Collapse
Affiliation(s)
- Aglaja Kopf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
13
|
Rezinciuc S, Tian Z, Wu S, Hengel S, Pasa-Tolic L, Smallwood HS. Mapping Influenza-Induced Posttranslational Modifications on Histones from CD8+ T Cells. Viruses 2020; 12:v12121409. [PMID: 33302437 PMCID: PMC7762524 DOI: 10.3390/v12121409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/23/2020] [Accepted: 12/02/2020] [Indexed: 12/25/2022] Open
Abstract
T cell function is determined by transcriptional networks that are regulated by epigenetic programming via posttranslational modifications (PTMs) to histone proteins and DNA. Bottom-up mass spectrometry (MS) can identify histone PTMs, whereas intact protein analysis by MS can detect species missed by bottom-up approaches. We used a novel approach of online two-dimensional liquid chromatography-tandem MS with high-resolution reversed-phase liquid chromatography (RPLC), alternating electron transfer dissociation (ETD) and collision-induced dissociation (CID) on precursor ions to maximize fragmentation of uniquely modified species. The first online RPLC separation sorted histone families, then RPLC or weak cation exchange hydrophilic interaction liquid chromatography (WCX-HILIC) separated species heavily clad in PTMs. Tentative identifications were assigned by matching proteoform masses to predicted theoretical masses that were verified with tandem MS. We used this innovative approach for histone-intact protein PTM mapping (HiPTMap) to identify and quantify proteoforms purified from CD8 T cells after in vivo influenza infection. Activation significantly altered PTMs following influenza infection, histone maps changed as T cells migrated to the site of infection, and T cells responding to secondary infections had significantly more transcription enhancing modifications. Thus, HiPTMap identified and quantified proteoforms and determined changes in CD8 T cell histone PTMs over the course of infection.
Collapse
Affiliation(s)
- Svetlana Rezinciuc
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Zhixin Tian
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (Z.T.); (S.W.); (S.H.); (L.P.-T.)
| | - Si Wu
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (Z.T.); (S.W.); (S.H.); (L.P.-T.)
| | - Shawna Hengel
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (Z.T.); (S.W.); (S.H.); (L.P.-T.)
| | - Ljiljana Pasa-Tolic
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (Z.T.); (S.W.); (S.H.); (L.P.-T.)
| | - Heather S. Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
- Children’s Foundation Research Institute, Memphis, TN 38105, USA
- Correspondence: ; Tel.: +1-(901)-448–3068
| |
Collapse
|
14
|
Mota TM, McCann CD, Danesh A, Huang SH, Magat DB, Ren Y, Leyre L, Bui TD, Rohwetter TM, Kovacs CM, Benko E, MacLaren L, Wimpelberg A, Cannon CM, Hardy WD, Safrit JT, Jones RB. Integrated Assessment of Viral Transcription, Antigen Presentation, and CD8 + T Cell Function Reveals Multiple Limitations of Class I-Selective Histone Deacetylase Inhibitors during HIV-1 Latency Reversal. J Virol 2020; 94:e01845-19. [PMID: 32051267 PMCID: PMC7163115 DOI: 10.1128/jvi.01845-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/04/2020] [Indexed: 12/17/2022] Open
Abstract
Clinical trials investigating histone deacetylase inhibitors (HDACi) to reverse HIV-1 latency aim to expose reservoirs in antiretroviral (ARV)-treated individuals to clearance by immune effectors, yet have not driven measurable reductions in the frequencies of infected cells. We therefore investigated the effects of the class I-selective HDACi nanatinostat and romidepsin on various blocks to latency reversal and elimination, including viral splicing, antigen presentation, and CD8+ T cell function. In ex vivo CD4+ T cells from ARV-suppressed individuals, both HDACi significantly induced viral transcription, but not splicing nor supernatant HIV-1 RNA. In an HIV-1 latency model using autologous CD8+ T cell clones as biosensors of antigen presentation, neither HDACi-treated CD4+ T cell condition induced clone degranulation. Both HDACi also impaired the function of primary CD8+ T cells in viral inhibition assays, with nanatinostat causing less impairment. These findings suggest that spliced or cell-free HIV-1 RNAs are more indicative of antigen expression than unspliced HIV-RNAs and may help to explain the limited abilities of HDACi to generate CD8+ T cell targets in vivoIMPORTANCE Antiretroviral (ARV) drug regimens suppress HIV-1 replication but are unable to cure infection. This leaves people living with HIV-1 burdened by a lifelong commitment to expensive daily medication. Furthermore, it has become clear that ARV therapy does not fully restore health, leaving individuals at elevated risk for cardiovascular disease, certain types of cancers, and neurocognitive disorders, as well as leaving them exposed to stigma. Efforts are therefore under way to develop therapies capable of curing infection. A key focus of these efforts has been on a class of drugs called histone deacetylase inhibitors (HDACi), which have the potential of exposing hidden reservoirs of HIV-1 to elimination by the immune system. Unfortunately, clinical trial results with HDACi have thus far been disappointing. In the current study, we integrate a number of experimental approaches to build a model that provides insights into the limited activity of HDACi in clinical trials and offers direction for future approaches.
Collapse
Affiliation(s)
- Talia M Mota
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Chase D McCann
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Ali Danesh
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Szu-Han Huang
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Dean B Magat
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Yanqin Ren
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Louise Leyre
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Tracy D Bui
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Thomas M Rohwetter
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| | | | | | - Lynsay MacLaren
- Research Department, Whitman-Walker Health, Washington, DC, USA
| | | | | | - W David Hardy
- Division of Infectious Disease, Johns Hopkins University School of Medicine, Washington, DC, USA
| | | | - R Brad Jones
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
15
|
Saeed MB, Record J, Westerberg LS. Two sides of the coin: Cytoskeletal regulation of immune synapses in cancer and primary immune deficiencies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:1-97. [DOI: 10.1016/bs.ircmb.2020.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
16
|
Yerinde C, Siegmund B, Glauben R, Weidinger C. Metabolic Control of Epigenetics and Its Role in CD8 + T Cell Differentiation and Function. Front Immunol 2019; 10:2718. [PMID: 31849941 PMCID: PMC6901948 DOI: 10.3389/fimmu.2019.02718] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
Epigenetic programs that control posttranslational modifications of histone proteins and DNA itself tightly regulate transcriptional networks determining the identity and function of CD8+ T cells. Chromatin-modifying enzymes such as histone acetyltransferases and deacetylases, represent key molecular determinants of the epigenetic imprinting of CD8+ T cells. The functions of these enzymes highly depend on the availability of key products of cellular metabolism pathways such as acetyl-CoA, NAD (Nicotinamide adenine dinucleotide) and SEM (S-adenosylmethionine), suggesting that there is a close crosstalk between the metabolic and the epigenetic regulation of CD8+ T cells. In this review, we will discuss the metabolic regulation of CD8+ T cell epigenetics during activation and differentiation. We will furthermore summarize how metabolic signals from the tumor microenvironment (TME) shape the epigenetic landscape of CD8+ T cells to better understand the mechanism underlying CD8+ T cell exhaustion in anti-tumor and anti-viral immunity, which might help to overcome limitations of current CD8+ T cell-based therapies.
Collapse
Affiliation(s)
- Cansu Yerinde
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rainer Glauben
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carl Weidinger
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Clinician Scientist Program, Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
17
|
Li C, Zhu B, Son Y, Wang Z, Jiang L, Xiang M, Ye Z, Beckermann KE, Wu Y, Jenkins J, Siska PJ, Vincent BG, Prakash Y, Peikert T, Edelson BT, Taneja R, Kaplan MH, Rathmell JC, Dong H, Hitosugi T, Sun J. The Transcription Factor Bhlhe40 Programs Mitochondrial Regulation of Resident CD8 + T Cell Fitness and Functionality. Immunity 2019; 51:491-507.e7. [PMID: 31533057 PMCID: PMC6903704 DOI: 10.1016/j.immuni.2019.08.013] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 04/13/2019] [Accepted: 08/14/2019] [Indexed: 12/28/2022]
Abstract
Tissue-resident memory CD8+ T (Trm) cells share core residency gene programs with tumor-infiltrating lymphocytes (TILs). However, the transcriptional, metabolic, and epigenetic regulation of Trm cell and TIL development and function is largely undefined. Here, we found that the transcription factor Bhlhe40 was specifically required for Trm cell and TIL development and polyfunctionality. Local PD-1 signaling inhibited TIL Bhlhe40 expression, and Bhlhe40 was critical for TIL reinvigoration following anti-PD-L1 blockade. Mechanistically, Bhlhe40 sustained Trm cell and TIL mitochondrial fitness and a functional epigenetic state. Building on these findings, we identified an epigenetic and metabolic regimen that promoted Trm cell and TIL gene signatures associated with tissue residency and polyfunctionality. This regimen empowered the anti-tumor activity of CD8+ T cells and possessed therapeutic potential even at an advanced tumor stage in mouse models. Our results provide mechanistic insights into the local regulation of Trm cell and TIL function.
Collapse
Affiliation(s)
- Chaofan Li
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Bibo Zhu
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Youngmin Son
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Zheng Wang
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Li Jiang
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Min Xiang
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhenqing Ye
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Kathryn E. Beckermann
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yue Wu
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - James Jenkins
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Peter J. Siska
- Internal Medicine III, University Hospital Regensburg, 93042 Regensburg, Germany,Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Y.S. Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Tobias Peikert
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Brian T. Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Reshma Taneja
- Department of Physiology, National University of Singapore, Singapore 117593
| | - Mark H. Kaplan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jeffrey C. Rathmell
- Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Taro Hitosugi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jie Sun
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
18
|
Cassioli C, Baldari CT. A Ciliary View of the Immunological Synapse. Cells 2019; 8:E789. [PMID: 31362462 PMCID: PMC6721628 DOI: 10.3390/cells8080789] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/15/2019] [Accepted: 07/25/2019] [Indexed: 12/28/2022] Open
Abstract
The primary cilium has gone from being a vestigial organelle to a crucial signaling hub of growing interest given the association between a group of human disorders, collectively known as ciliopathies, and defects in its structure or function. In recent years many ciliogenesis proteins have been observed at extraciliary sites in cells and likely perform cilium-independent functions ranging from regulation of the cytoskeleton to vesicular trafficking. Perhaps the most striking example is the non-ciliated T lymphocyte, in which components of the ciliary machinery are repurposed for the assembly and function of the immunological synapse even in the absence of a primary cilium. Furthermore, the specialization traits described at the immunological synapse are similar to those seen in the primary cilium. Here, we review common regulators and features shared by the immunological synapse and the primary cilium that document the remarkable homology between these structures.
Collapse
Affiliation(s)
- Chiara Cassioli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| |
Collapse
|
19
|
Cao Y, Banks DA, Mattei AM, Riddick AT, Reed KM, Zhang AM, Pickering ES, Hinton SD. Pseudophosphatase MK-STYX Alters Histone Deacetylase 6 Cytoplasmic Localization, Decreases Its Phosphorylation, and Increases Detyrosination of Tubulin. Int J Mol Sci 2019; 20:ijms20061455. [PMID: 30909412 PMCID: PMC6470616 DOI: 10.3390/ijms20061455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 12/14/2022] Open
Abstract
The catalytically inactive mitogen-activated protein (MAP) kinase phosphatase, MK-STYX (MAPK (mitogen-activated protein kinase) phosphoserine/threonine/tyrosine-binding protein) interacts with the stress granule nucleator G3BP-1 (Ras-GAP (GTPase-activating protein) SH3 (Src homology 3) domain-binding protein-1), and decreases stress granule (stalled mRNA) formation. Histone deacetylase isoform 6 (HDAC6) also binds G3BP-1 and serves as a major component of stress granules. The discovery that MK-STYX and HDAC6 both interact with G3BP-1 led us to investigate the effects of MK-STYX on HDAC6 dynamics. In control HEK/293 cells, HDAC6 was cytosolic, as expected, and formed aggregates under conditions of stress. In contrast, in cells overexpressing MK-STYX, HDAC6 was both nuclear and cytosolic and the number of stress-induced aggregates significantly decreased. Immunoblots showed that MK-STYX decreases HDAC6 serine phosphorylation, protein tyrosine phosphorylation, and lysine acetylation. HDAC6 is known to regulate microtubule dynamics to form aggregates. MK-STYX did not affect the organization of microtubules, but did affect their post-translational modification. Tubulin acetylation was increased in the presence of MK-STYX. In addition, the detyrosination of tubulin was significantly increased in the presence of MK-STYX. These findings show that MK-STYX decreases the number of HDAC6-containing aggregates and alters their localization, sustains microtubule acetylation, and increases detyrosination of microtubules, implicating MK-STYX as a signaling molecule in HDAC6 activity.
Collapse
Affiliation(s)
- Yuming Cao
- Department of Biology, Integrated Science Center, College of William and Mary, Williamsburg, VA 23185, USA.
| | - Dallas A Banks
- Department of Biology, Integrated Science Center, College of William and Mary, Williamsburg, VA 23185, USA.
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA.
| | - Andrew M Mattei
- Department of Biology, Integrated Science Center, College of William and Mary, Williamsburg, VA 23185, USA.
| | - Alexys T Riddick
- Department of Biology, Integrated Science Center, College of William and Mary, Williamsburg, VA 23185, USA.
| | - Kirstin M Reed
- Department of Biology, Integrated Science Center, College of William and Mary, Williamsburg, VA 23185, USA.
| | - Ashley M Zhang
- Department of Biology, Integrated Science Center, College of William and Mary, Williamsburg, VA 23185, USA.
| | - Emily S Pickering
- Department of Biology, Integrated Science Center, College of William and Mary, Williamsburg, VA 23185, USA.
| | - Shantá D Hinton
- Department of Biology, Integrated Science Center, College of William and Mary, Williamsburg, VA 23185, USA.
| |
Collapse
|
20
|
Aurora A controls CD8 + T cell cytotoxic activity and antiviral response. Sci Rep 2019; 9:2211. [PMID: 30778113 PMCID: PMC6379542 DOI: 10.1038/s41598-019-38647-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/28/2018] [Indexed: 12/05/2022] Open
Abstract
Aurora A is a serine/threonine kinase whose role in cell cycle progression and tumour generation has been widely studied. Recent work has revealed an unexpected function for Aurora A during CD4+ T cell activation and, also, in graft versus host disease development. However, it remains unknown whether Aurora A is involved in CD8+ T cell effector function and in cytotoxic T lymphocyte-mediated antiviral response. Here, we show that Aurora A chemical inhibition leads to an impairment of both the peptide-specific cytotoxicity and the degranulation activity of CD8+ T cells. This finding was similarly proven for both mice and human CD8+ CTL activity. As a result of Aurora A blockade, we detected a reduction in the expression induced by T cell activation of genes classically related to the effector function of cytotoxic T lymphocytes such as granzyme B or perforin1. Finally, we have found that Aurora A is necessary for CD8+ T cell-mediated antiviral response, in an in vivo model of vaccinia virus infection. Thus, we can conclude that Aurora A activity is, indeed, needed for the proper effector function of cytotoxic T lymphocytes and for their activity against viral threats.
Collapse
|
21
|
Abstract
The differentiation of T helper cell subsets and their acquisition of effector functions are accompanied by changes in gene expression programmes, which in part are regulated and maintained by epigenetic processes. Histone deacetylases (HDACs) and histone acetyltransferases (HATs) are key epigenetic regulators that function by mediating dynamic changes in the acetylation of histones at lysine residues. In addition, many non-histone proteins are also acetylated, and reversible acetylation affects their functional properties, demonstrating that HDACs mediate effects beyond the epigenetic regulation of gene expression. In this Review, we discuss studies revealing that HDACs are key regulators of CD4+ T cell-mediated immunity in mice and humans and that HDACs are promising targets in T cell-mediated immune diseases. Finally, we discuss unanswered questions and future research directions to promote the concept that isoform-selective HDAC inhibitors might broaden the clinical application of HDAC inhibitors beyond their current use in certain types of cancer.
Collapse
Affiliation(s)
- Wilfried Ellmeier
- Division of Immunobiology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Christian Seiser
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Moreno-Gonzalo O, Mayor F, Sánchez-Madrid F. HDAC6 at Crossroads of Infection and Innate Immunity. Trends Immunol 2018; 39:591-595. [DOI: 10.1016/j.it.2018.05.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/07/2018] [Accepted: 05/28/2018] [Indexed: 11/28/2022]
|
23
|
Martín-Cófreces NB, Sánchez-Madrid F. Sailing to and Docking at the Immune Synapse: Role of Tubulin Dynamics and Molecular Motors. Front Immunol 2018; 9:1174. [PMID: 29910809 PMCID: PMC5992405 DOI: 10.3389/fimmu.2018.01174] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/11/2018] [Indexed: 12/17/2022] Open
Abstract
The different cytoskeleton systems and their connecting molecular motors move vesicles and intracellular organelles to shape cells. Polarized cells with specialized functions display an exquisite spatio-temporal regulation of both cytoskeletal and organelle arrangements that support their specific tasks. In particular, T cells rapidly change their shape and cellular function through the establishment of cell surface and intracellular polarity in response to a variety of cues. This review focuses on the contribution of the microtubule-based dynein/dynactin motor complex, the tubulin and actin cytoskeletons, and different organelles to the formation of the antigen-driven immune synapse.
Collapse
Affiliation(s)
- Noa Beatriz Martín-Cófreces
- Servicio de Inmunología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
24
|
Abstract
The oncogene HDAC6 controls numerous cell processes that are related to tumorigenesis and metastasis, and has recently arisen as a target to treat malignancies. The ERK cascade is a classic pathway driving oncogenesis, and the components of this pathway are either highly mutated in cancers or are vital in cancer's pathological activity. The interactions between these important components of tumor proliferation have been examined, and our research has demonstrated that they regulate each other as evidenced by different posttranslational modifications. Preclinical evidence also supports clinical trials cotargeting these two pathways, which may provide better efficacy than single treatment. Furthermore, HDAC6 and ERK both participate in the regulation of T cell maturation and may have implications on the functions of immune cells. This leads to the possibility of connecting HDAC6 and ERK to immunotherapy. In this review, we summarize the published studies about the interaction of HDAC6 and ERK cascade and their relationship to cancers. We also include the association of HDAC6 and ERK to immune system and discuss the plausibility of linking these to immunotherapy.
Collapse
Affiliation(s)
- Jheng-Yu Wu
- Department of Oncology, Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, Michigan 48201.,Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Niko Moses
- Department of Oncology, Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, Michigan 48201.,Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Wenlong Bai
- Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Xiaohong Mary Zhang
- Department of Oncology, Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, Michigan 48201.,Wayne State University School of Medicine, Detroit, Michigan 48201
| |
Collapse
|
25
|
McCaw TR, Randall TD, Forero A, Buchsbaum DJ. Modulation of antitumor immunity with histone deacetylase inhibitors. Immunotherapy 2017; 9:1359-1372. [PMID: 29185390 PMCID: PMC6077764 DOI: 10.2217/imt-2017-0134] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/30/2017] [Indexed: 01/02/2023] Open
Abstract
Histone deacetylase inhibitors possess a broad array of antitumor activities; however, their net impact on the evolving antitumor immune response is highly dependent on the inhibitors used and the histone deacetylases they target. Herein, we sequentially focus on each stage of the antitumor immune response - from dendritic cell activation and migration, antigen uptake and presentation, T-cell activation and differentiation and the enactment of antitumor effector functions within the tumor microenvironment. In particular, we will discuss how various inhibitors have different effects depending on cellular activation, experimental design and specific histone deacetylases being targeted - and how these changes impact the outcome of an antitumor immune response. At last, we consider the impact these inhibitors may have on T-cell exhaustion and implications for combination with other immunomodulating therapies.
Collapse
Affiliation(s)
- Tyler R McCaw
- Department of Medicine, Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA, 35233
| | - Troy D Randall
- Department of Medicine, Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA, 35233
| | - Andres Forero
- Department of Medicine, Division of Hematology & Oncology, University of Alabama at Birmingham, Birmingham, AL, USA, 35233
| | - Donald J Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA, 35233
| |
Collapse
|
26
|
Villarroya-Beltri C, Guerra S, Sánchez-Madrid F. ISGylation - a key to lock the cell gates for preventing the spread of threats. J Cell Sci 2017; 130:2961-2969. [PMID: 28842471 DOI: 10.1242/jcs.205468] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Interferon stimulated gene 15 (ISG15) is an ubiquitin-like protein whose expression and conjugation to targets (ISGylation) is induced by infection, interferon (IFN)-α and -β, ischemia, DNA damage and aging. Attention has historically focused on the antiviral effects of ISGylation, which blocks the entry, replication or release of different intracellular pathogens. However, recently, new functions of ISGylation have emerged that implicate it in multiple cellular processes, such as DNA repair, autophagy, protein translation and exosome secretion. In this Review, we discuss the induction and conjugation of ISG15, as well as the functions of ISGylation in the prevention of infections and in cancer progression. We also offer a novel perspective with regard to the latest findings on this pathway, with special attention to the role of ISGylation in the inhibition of exosome secretion, which is mediated by fusion of multivesicular bodies with lysosomes. Finally, we propose that under conditions of stress or infection, ISGylation acts as a defense mechanism to inhibit normal protein translation by modifying protein kinase R (PKR, also known as EIF2AK2), while any newly synthesized proteins are being tagged and thus marked as potentially dangerous. Then, the endosomal system is re-directed towards protein degradation at the lysosome, to effectively 'lock' the cell gates and thus prevent the spread of pathogens, prions and deleterious aggregates through exosomes.
Collapse
Affiliation(s)
- Carolina Villarroya-Beltri
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.,Immunology Service, Hospital de la Princesa, Universidad Autónoma de Madrid, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Susana Guerra
- Preventive Medicine Department, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Francisco Sánchez-Madrid
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain .,Immunology Service, Hospital de la Princesa, Universidad Autónoma de Madrid, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
27
|
Zheng K, Jiang Y, He Z, Kitazato K, Wang Y. Cellular defence or viral assist: the dilemma of HDAC6. J Gen Virol 2017; 98:322-337. [PMID: 27959772 DOI: 10.1099/jgv.0.000679] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) is a unique cytoplasmic deacetylase that regulates various important biological processes by preventing protein aggregation and deacetylating different non-histone substrates including tubulin, heat shock protein 90, cortactin, retinoic acid inducible gene I and β-catenin. Growing evidence has indicated a dual role for HDAC6 in viral infection and pathogenesis: HDAC6 may represent a host defence mechanism against viral infection by modulating microtubule acetylation, triggering antiviral immune response and stimulating protective autophagy, or it may be hijacked by the virus to enhance proinflammatory response. In this review, we will highlight current data illustrating the complexity and importance of HDAC6 in viral pathogenesis. We will summarize the structure and functional specificity of HDAC6, and its deacetylase- and ubiquitin-dependent activity in key cellular events in response to virus infection. We will also discuss how HDAC6 exerts its direct or indirect histone modification ability in viral lytic-latency switch.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Pharmacy, School of Medicine, Shenzhen University, Shenzhen 518060, PR China.,College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| | - Yingchun Jiang
- Department of Pharmacy, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Zhendan He
- Department of Pharmacy, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Kaio Kitazato
- Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Yifei Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| |
Collapse
|
28
|
Bustos-Morán E, Blas-Rus N, Martin-Cófreces NB, Sánchez-Madrid F. Microtubule-associated protein-4 controls nanovesicle dynamics and T cell activation. J Cell Sci 2017; 130:1217-1223. [PMID: 28209780 DOI: 10.1242/jcs.199042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/07/2017] [Indexed: 01/06/2023] Open
Abstract
The immune synapse (IS) is a specialized structure formed at the contact area between T lymphocytes and antigen-presenting cells (APCs) that is essential for the adaptive immune response. Proper T cell activation requires its polarization towards the APC, which is highly dependent on the tubulin cytoskeleton. Microtubule-associated protein-4 (MAP4) is a microtubule (MT)-stabilizing protein that controls MTs in physiological processes, such as cell division, migration, vesicular transport or primary cilia formation. In this study, we assessed the role of MAP4 in T cell activation. MAP4 decorates the pericentrosomal area and MTs of the T cell, and it is involved in MT detyrosination and stable assembly in response to T cell activation. In addition, MAP4 prompts the timely translocation of the MT-organizing center (MTOC) towards the IS and the dynamics of signaling nanovesicles that sustains T cell activation. However, MAP4 acts as a negative regulator of other T cell activation-related signals, including diacylglycerol (DAG) production and IL2 secretion. Our data indicate that MAP4 acts as a checkpoint molecule that balances positive and negative hallmarks of T cell activation.
Collapse
Affiliation(s)
- Eugenio Bustos-Morán
- Laboratory of Intercellular communication, Fundación CNIC, Madrid 28029, Spain.,Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid 28006, Spain
| | - Noelia Blas-Rus
- Laboratory of Intercellular communication, Fundación CNIC, Madrid 28029, Spain.,Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid 28006, Spain
| | - Noa Beatriz Martin-Cófreces
- Laboratory of Intercellular communication, Fundación CNIC, Madrid 28029, Spain.,Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid 28006, Spain.,CIBER Cardiovascular, ISCIII, Madrid, 28029, Spain
| | - Francisco Sánchez-Madrid
- Laboratory of Intercellular communication, Fundación CNIC, Madrid 28029, Spain .,Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid 28006, Spain.,CIBER Cardiovascular, ISCIII, Madrid, 28029, Spain
| |
Collapse
|
29
|
Nunez-Andrade N, Sanchez-Madrid F, Martin-Cofreces NB. HDAC6 is a Regulator of CTL Function through Control of Lytic Granule Dynamics. ACTA ACUST UNITED AC 2016; 5. [PMID: 27595053 DOI: 10.4172/2168-9431.1000143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Viral infections involve specific stress exposure that can influence the quality and average lifespan of an organism. The immune system acts through virus clearance from the organism. Many aspects of immune cells accounting for this response are still under study. Here, we review recent aspects of the molecular mechanisms involved in the delivery of the lethal hit by Cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Norman Nunez-Andrade
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid, Spain.,Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, Spain
| | - Francisco Sanchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid, Spain.,Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, Spain
| | - Noa Beatriz Martin-Cofreces
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid, Spain.,Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, Spain
| |
Collapse
|
30
|
Bustos-Morán E, Blas-Rus N, Martín-Cófreces NB, Sánchez-Madrid F. Orchestrating Lymphocyte Polarity in Cognate Immune Cell-Cell Interactions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 327:195-261. [PMID: 27692176 DOI: 10.1016/bs.ircmb.2016.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immune synapse (IS) is a specialized structure established between different immune cells that fulfills several functions, including a role as a communication bridge. This intimate contact between a T cell and an antigen-presenting cell promotes the proliferation and differentiation of lymphocytes involved in the contact. T-cell activation requires the specific triggering of the T-cell receptor (TCR), which promotes the activation of different signaling pathways inducing the polarization of the T cell. During this process, different adhesion and signaling receptors reorganize at specialized membrane domains, concomitantly to the polarization of the tubulin and actin cytoskeletons, forming stable polarization platforms. The centrosome also moves toward the IS, driving the movement of different organelles, such as the biosynthetic, secretory, degrading machinery, and mitochondria, to sustain T-cell activation. A proper orchestration of all these events is essential for T-cell effector functions and the accomplishment of a complete immune response.
Collapse
Affiliation(s)
- Eugenio Bustos-Morán
- Vascular Pathophysiology Area, Spanish National Center of Cardiovascular Research (CNIC), Madrid, Spain
| | - Noelia Blas-Rus
- Department of Immunology, La Princesa Hospital, Autonomus University of Madrid (UAM), Health Research Institute of Princesa Hospital (ISS-IP), Madrid, Spain
| | - Noa Beatriz Martín-Cófreces
- Vascular Pathophysiology Area, Spanish National Center of Cardiovascular Research (CNIC), Madrid, Spain.,Department of Immunology, La Princesa Hospital, Autonomus University of Madrid (UAM), Health Research Institute of Princesa Hospital (ISS-IP), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Vascular Pathophysiology Area, Spanish National Center of Cardiovascular Research (CNIC), Madrid, Spain.,Department of Immunology, La Princesa Hospital, Autonomus University of Madrid (UAM), Health Research Institute of Princesa Hospital (ISS-IP), Madrid, Spain
| |
Collapse
|