1
|
Li Z, Yang J, Ma R, Xie S, Wang D, Quan R, Wen X, Liu J, Song J. Seneca Valley virus 3C protease cleaves HDAC4 to antagonize type I interferon signaling. J Virol 2025; 99:e0217624. [PMID: 39927774 PMCID: PMC11915795 DOI: 10.1128/jvi.02176-24] [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: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 02/11/2025] Open
Abstract
Seneca Valley virus (SVV) is a newly identified pathogen that poses a notable threat to the global pig industry. SVV has evolved multiple strategies to evade host antiviral innate immune responses. However, the underlying molecular mechanisms have not yet been fully elucidated. Histone deacetylases (HDACs) have been shown to function as host antiviral innate immune factors. In this study, we examined the mechanisms underlying SVV evasion of host innate immunity and found that SVV infection induced degradation and cleavage of HDAC4. Ectopic expression of HDAC4 suppressed SVV replication, whereas siRNA-mediated knockdown of HDAC4 enhanced SVV replication. Further studies showed that the viral 3C protease (3Cpro) degraded HDAC4 in a protease activity- and caspase pathway-dependent manner. In addition, 3Cpro cleaved HDAC4 at Q599, which blocked its ability to limit viral replication. We also found that HDAC4 interacted with the SVV viral RNA-dependent RNA polymerase 3D and induced its proteasomal degradation. The cleaved HDAC4 products did not block SVV replication or induce 3D degradation and did not induce type I interferon (IFN) activation and expression of IFN-stimulated genes (ISGs). Collectively, these findings identified HDAC4 as an antiviral factor with effects against SVV infection and provided mechanistic insights into how SVV 3Cpro antagonizes its function, which has implications for viral evasion of innate immunity. IMPORTANCE Seneca Valley virus (SVV) is an emerging pathogen that causes vesicular disease in pigs and poses a threat to the pork industry. Histone deacetylases (HDACs) are important in the regulation of innate immunity. However, little is known about their roles in SVV infection. Our results revealed HDAC4 as an anti-SVV infection factor that targets the viral RNA-dependent RNA polymerase, 3D, for degradation. The SVV proteinase 3Cpro targets HDAC4 for degradation and cleavage, and cleavage of HDAC4 abrogated its antiviral effect. HDAC4 promotes type I interferon (IFN) signaling, and SVV 3Cpro-mediated cleavage of HDAC4 antagonized induction of type I IFN and interferon-stimulated genes (ISGs). Our findings reveal a novel molecular mechanism by which SVV 3Cpro counteracts type I IFN signaling by targeting HDAC4.
Collapse
Affiliation(s)
- Zijian Li
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Department of Preventive Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Jingjing Yang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Department of Preventive Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Ruiyi Ma
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Shijie Xie
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Dan Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Rong Quan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Xuexia Wen
- Department of Preventive Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Jue Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiangwei Song
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| |
Collapse
|
2
|
Saha A, Ganguly A, Kumar A, Srivastava N, Pathak R. Harnessing Epigenetics: Innovative Approaches in Diagnosing and Combating Viral Acute Respiratory Infections. Pathogens 2025; 14:129. [PMID: 40005506 PMCID: PMC11858160 DOI: 10.3390/pathogens14020129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Acute respiratory infections (ARIs) caused by viruses such as SARS-CoV-2, influenza viruses, and respiratory syncytial virus (RSV), pose significant global health challenges, particularly for the elderly and immunocompromised individuals. Substantial evidence indicates that acute viral infections can manipulate the host's epigenome through mechanisms like DNA methylation and histone modifications as part of the immune response. These epigenetic alterations can persist beyond the acute phase, influencing long-term immunity and susceptibility to subsequent infections. Post-infection modulation of the host epigenome may help distinguish infected from uninfected individuals and predict disease severity. Understanding these interactions is crucial for developing effective treatments and preventive strategies for viral ARIs. This review highlights the critical role of epigenetic modifications following viral ARIs in regulating the host's innate immune defense mechanisms. We discuss the implications of these modifications for diagnosing, preventing, and treating viral infections, contributing to the advancement of precision medicine. Recent studies have identified specific epigenetic changes, such as hypermethylation of interferon-stimulated genes in severe COVID-19 cases, which could serve as biomarkers for early detection and disease progression. Additionally, epigenetic therapies, including inhibitors of DNA methyltransferases and histone deacetylases, show promise in modulating the immune response and improving patient outcomes. Overall, this review provides valuable insights into the epigenetic landscape of viral ARIs, extending beyond traditional genetic perspectives. These insights are essential for advancing diagnostic techniques and developing innovative treatments to address the growing threat of emerging viruses causing ARIs globally.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar 814152, India;
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India;
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
3
|
Liu D, Howard TR, Cristea IM. ATAC-seq for Characterizing Host and Pathogen Genome Accessibility During Virus Infection. Methods Mol Biol 2025; 2866:111-122. [PMID: 39546200 DOI: 10.1007/978-1-0716-4192-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Chromatin regulation provides a mechanism through which cells dynamically and rapidly regulate their gene expression profiles, playing a pivotal role in diverse biological processes and disease states. The Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) is a method that enables genome-wide detection of accessible chromatin regions, providing information on nucleosome positioning and the epigenetic regulation of the chromatin structure. ATAC-seq has been used in various biological contexts, and several reports have demonstrated its application to studying infections with viral or bacterial pathogens. The ability to characterize changes in viral or bacterial genome accessibility during infections provides insights into both pathogen replication and host defense mechanisms. Viral genomes undergo dynamic changes in their structural landscape to facilitate replication and evade host immune responses. Additionally, host cells encode DNA sensors, which are specialized proteins that bind to viral genomes to initiate innate immune responses and sometimes, to suppress viral gene expression. ATAC-seq enables the systematic detection of key structural changes on the viral genome mediated by either viral or host proteins, offering mechanistic insights into virus-host interactions. Here, we describe an ATAC-seq method optimized for studying changes in chromatin accessibility in both host and viral genomes. We have previously applied this method to demonstrate a systematic decrease in the genome accessibility of herpes simplex virus type I (HSV-1) enabled by a host antiviral factor, the interferon-gamma inducible protein 16 (IFI16) during infection of human fibroblasts. This protocol can be adapted to various biological contexts involving the introduction of foreign DNA, making it a valuable tool for a broad range of research endeavors.
Collapse
Affiliation(s)
- Dawei Liu
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Timothy R Howard
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
4
|
Raouf YS, Moreno-Yruela C. Slow-Binding and Covalent HDAC Inhibition: A New Paradigm? JACS AU 2024; 4:4148-4161. [PMID: 39610753 PMCID: PMC11600154 DOI: 10.1021/jacsau.4c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024]
Abstract
The dysregulated post-translational modification of proteins is an established hallmark of human disease. Through Zn2+-dependent hydrolysis of acyl-lysine modifications, histone deacetylases (HDACs) are key regulators of disease-implicated signaling pathways and tractable drug targets in the clinic. Early targeting of this family of 11 enzymes (HDAC1-11) afforded a first generation of broadly acting inhibitors with medicinal applications in oncology, specifically in cutaneous and peripheral T-cell lymphomas and in multiple myeloma. However, first-generation HDAC inhibitors are often associated with weak-to-modest patient benefits, dose-limited efficacies, pharmacokinetic liabilities, and recurring clinical toxicities. Alternative inhibitor design to target single enzymes and avoid toxic Zn2+-binding moieties have not overcome these limitations. Instead, recent literature has seen a shift toward noncanonical mechanistic approaches focused on slow-binding and covalent inhibition. Such compounds hold the potential of improving the pharmacokinetic and pharmacodynamic profiles of HDAC inhibitors through the extension of the drug-target residence time. This perspective aims to capture this emerging paradigm and discuss its potential to improve the preclinical/clinical outlook of HDAC inhibitors in the coming years.
Collapse
Affiliation(s)
- Yasir S. Raouf
- Department
of Chemistry, United Arab Emirates University, P.O. Box No. 15551 Al Ain, UAE
| | - Carlos Moreno-Yruela
- Laboratory
of Chemistry and Biophysics of Macromolecules (LCBM), Institute of
Chemical Sciences and Engineering (ISIC), School of Basic Sciences, École Polytechnique Fédérale
de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
5
|
Zeba A, Rajalingam A, Sekar K, Ganjiwale A. Machine learning-based gene expression biomarkers to distinguish Zika and Dengue virus infections: implications for diagnosis. Virusdisease 2024; 35:446-461. [PMID: 39464736 PMCID: PMC11502647 DOI: 10.1007/s13337-024-00885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 07/19/2024] [Indexed: 10/29/2024] Open
Abstract
Zika virus (ZIKV) and Dengue virus (DENV) infections cause severe disease in humans and are significant socio-economic burden worldwide. These flavivirus infections are difficult to diagnose serologically due to antigenic overlap. The phylogenetic analysis shows that ZIKV clusters with DENVs at a higher node of the phylogenetic tree with significant genomic and structural similarity. Our study aims to identify gene biomarkers for the classification of Dengue and Zika viral infections using machine learning algorithms and bioinformatics analysis. The gene expression count matrix for single-cell RNA sequencing dataset GSE110496 was analyzed using binary classifiers, namely Logistic regression, Support Vector Machines, Random Forest, and Decision trees. The GSE110496 dataset represents a unique study of the transcriptional and translational dynamics of DENV and ZIKV infections at 4-, 12-, 24-, and 48-h time points for human hepatoma (Huh7) cells. Out of which 24-h time point has been analyzed in this study, at the optimal threshold of viral molecules. Feature selection was performed using two different approaches Random Forest Classifier (RFC) for gene ranking and Recursive Feature Elimination (RFE). Out of which RFE, showed more accuracy and precision. The classification accuracy of 89.4% and the precision of 90% were obtained using selected 10 gene features. SCY1 Like Pseudokinase 3 (SCYL3), Chromosome 1 Open Reading Frame 112 (C1orf112), Complement factor H (CFH), Heme-binding protein 1 (HEBP1), Cadherin 1 (CDH1), Nibrin (NBN), Histone deacetylase 5 (HDAC5), nuclear receptor subfamily 0, group B, member 2 (NR0B2), Annexin A9 (ANXA9) and Alcohol dehydrogenase 6 (ADH6) are the proposed gene biomarkers in this study. The functional analysis of the reported biomarkers was performed using KEGG and GO with the WEB-based Gene SeT AnaLysis Toolkit (WebGestalt). The relationship of the selected biomarkers with DENV and ZIKV infections analyzed using a gene-gene interaction network showed important interactions for viral entry, replication, translation, and metabolic pathways. These biomarkers are potential diagnostic markers for DENV and ZIKV infections based on machine learning analysis and need further experimental validation. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s13337-024-00885-8.
Collapse
Affiliation(s)
- Ayesha Zeba
- Department of Life Science, Bangalore University, Bangalore, Karnataka 560056 India
| | - Aruna Rajalingam
- Department of Life Science, Bangalore University, Bangalore, Karnataka 560056 India
| | - Kanagaraj Sekar
- Laboratory for Structural Biology and Bio-Computing, Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, Karnataka 560012 India
| | - Anjali Ganjiwale
- Department of Life Science, Bangalore University, Bangalore, Karnataka 560056 India
| |
Collapse
|
6
|
Raouf YS. Targeting histone deacetylases: Emerging applications beyond cancer. Drug Discov Today 2024; 29:104094. [PMID: 38997001 DOI: 10.1016/j.drudis.2024.104094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/25/2024] [Accepted: 07/04/2024] [Indexed: 07/14/2024]
Abstract
Histone deacetylases (HDACs) are a special class of hydrolase enzymes, which through epigenetic control of cellular acetylation, play regulatory roles in various processes including chromatin packing, cytokine signaling, and gene expression. Widespread influence on cell function has implicated dysregulated HDAC activity in human disease. While traditionally an oncology target, in the past decade, there has been a notable rise in inhibition strategies within several therapeutic areas beyond cancer. This review highlights advances in four of these indications, neurodegenerative disease, metabolic disorders, cardiovascular disease, and viral infections, focusing on the role of deacetylases in disease, small molecule drug discovery, and clinical progress.
Collapse
Affiliation(s)
- Yasir S Raouf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, P.O. Box 15551, United Arab Emirates.
| |
Collapse
|
7
|
Shi MQ, Xu Y, Fu X, Pan DS, Lu XP, Xiao Y, Jiang YZ. Advances in targeting histone deacetylase for treatment of solid tumors. J Hematol Oncol 2024; 17:37. [PMID: 38822399 PMCID: PMC11143662 DOI: 10.1186/s13045-024-01551-8] [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: 01/03/2024] [Accepted: 04/27/2024] [Indexed: 06/03/2024] Open
Abstract
Histone deacetylase (HDAC) serves as a critical molecular regulator in the pathobiology of various malignancies and have garnered attention as a viable target for therapeutic intervention. A variety of HDAC inhibitors (HDACis) have been developed to target HDACs. Many preclinical studies have conclusively demonstrated the antitumor effects of HDACis, whether used as monotherapy or in combination treatments. On this basis, researchers have conducted various clinical studies to evaluate the potential of selective and pan-HDACis in clinical settings. In our work, we extensively summarized and organized current clinical trials, providing a comprehensive overview of the current clinical advancements in targeting HDAC therapy. Furthermore, we engaged in discussions about several clinical trials that did not yield positive outcomes, analyzing the factors that led to their lack of anticipated therapeutic effectiveness. Apart from the experimental design factors, issues such as toxicological side effects, tumor heterogeneity, and unexpected off-target effects also contributed to these less-than-expected results. These challenges have naturally become significant barriers to the application of HDACis. Despite these challenges, we believe that advancements in HDACi research and improvements in combination therapies will pave the way or lead to a broad and hopeful future in the treatment of solid tumors.
Collapse
Affiliation(s)
- Mu-Qi Shi
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ying Xu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xin Fu
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen, 518055, People's Republic of China
| | - De-Si Pan
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen, 518055, People's Republic of China
| | - Xian-Ping Lu
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen, 518055, People's Republic of China
| | - Yi Xiao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
8
|
Lu Y, Zhao Y, Gao C, Suresh S, Men J, Sawyers A, Smith GL. HDAC5 enhances IRF3 activation and is targeted for degradation by protein C6 from orthopoxviruses including Monkeypox virus and Variola virus. Cell Rep 2024; 43:113788. [PMID: 38461415 PMCID: PMC11650635 DOI: 10.1016/j.celrep.2024.113788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/18/2023] [Accepted: 01/26/2024] [Indexed: 03/12/2024] Open
Abstract
Histone deacetylases (HDACs) regulate gene expression and innate immunity. Previously, we showed that HDAC5 is degraded during Vaccinia virus (VACV) infection and is a restriction factor for VACV and herpes simplex virus type 1. Here, we report that HDAC5 promotes interferon regulatory factor 3 (IRF3) activation downstream of Toll-IL-1 receptor (TIR) domain-containing adaptor molecule-1 or Sendai virus-mediated stimulation without requiring HDAC activity. Loss of HDAC5-mediated IRF3 activation is restored by re-introduction of HDAC5 but not HDAC1 or HDAC4. The antiviral activity of HDAC5 is antagonized by VACV protein C6 and orthologs from the orthopoxviruses cowpox, rabbitpox, camelpox, monkeypox, and variola. Infection by many of these viruses induces proteasomal degradation of HDAC5, and expression of C6 alone can induce HDAC5 degradation. Mechanistically, C6 binds to the dimerization domain of HDAC5 and prevents homodimerization and heterodimerization with HDAC4. Overall, this study describes HDAC5 as a positive regulator of IRF3 activation and provides mechanistic insight into how the poxviral protein C6 binds to HDAC5 to antagonize its function.
Collapse
Affiliation(s)
- Yongxu Lu
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK; Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; Chinese Academy of Medical Sciences-Oxford Institute, University of Oxford, Oxford, UK.
| | - Yiqi Zhao
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK; Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; Chinese Academy of Medical Sciences-Oxford Institute, University of Oxford, Oxford, UK
| | - Chen Gao
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Shreehari Suresh
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Jinghao Men
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Amelia Sawyers
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK; Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; The Pirbright Institute, Surrey, UK; Chinese Academy of Medical Sciences-Oxford Institute, University of Oxford, Oxford, UK.
| |
Collapse
|
9
|
Trionfetti F, Montaldo C, Caiello I, Bontempi G, Terri M, Tiberi M, Marchant V, Domenici A, Menè P, Cordani M, Zwergel C, Prencipe G, Ruiz-Ortega M, Valente S, Mai A, Tripodi M, Strippoli R. Mechanisms of mesothelial cell response to viral infections: HDAC1-3 inhibition blocks poly(I:C)-induced type I interferon response and modulates the mesenchymal/inflammatory phenotype. Front Cell Infect Microbiol 2024; 14:1308362. [PMID: 38476167 PMCID: PMC10927979 DOI: 10.3389/fcimb.2024.1308362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/24/2024] [Indexed: 03/14/2024] Open
Abstract
Infectious peritonitis is a leading cause of peritoneal functional impairment and a primary factor for therapy discontinuation in peritoneal dialysis (PD) patients. Although bacterial infections are a common cause of peritonitis episodes, emerging evidence suggests a role for viral pathogens. Toll-like receptors (TLRs) specifically recognize conserved pathogen-associated molecular patterns (PAMPs) from bacteria, viruses, and fungi, thereby orchestrating the ensuing inflammatory/immune responses. Among TLRs, TLR3 recognizes viral dsRNA and triggers antiviral response cascades upon activation. Epigenetic regulation, mediated by histone deacetylase (HDAC), has been demonstrated to control several cellular functions in response to various extracellular stimuli. Employing epigenetic target modulators, such as epidrugs, is a current therapeutic option in several cancers and holds promise in treating viral diseases. This study aims to elucidate the impact of TLR3 stimulation on the plasticity of human mesothelial cells (MCs) in PD patients and to investigate the effects of HDAC1-3 inhibition. Treatment of MCs from PD patients with the TLR3 agonist polyinosinic:polycytidylic acid (Poly(I:C)), led to the acquisition of a bona fide mesothelial-to-mesenchymal transition (MMT) characterized by the upregulation of mesenchymal genes and loss of epithelial-like features. Moreover, Poly(I:C) modulated the expression of several inflammatory cytokines and chemokines. A quantitative proteomic analysis of MCs treated with MS-275, an HDAC1-3 inhibitor, unveiled altered expression of several proteins, including inflammatory cytokines/chemokines and interferon-stimulated genes (ISGs). Treatment with MS-275 facilitated MMT reversal and inhibited the interferon signature, which was associated with reduced STAT1 phosphorylation. However, the modulation of inflammatory cytokine/chemokine production was not univocal, as IL-6 and CXCL8 were augmented while TNF-α and CXCL10 were decreased. Collectively, our findings underline the significance of viral infections in acquiring a mesenchymal-like phenotype by MCs and the potential consequences of virus-associated peritonitis episodes for PD patients. The observed promotion of MMT reversal and interferon response inhibition by an HDAC1-3 inhibitor, albeit without a general impact on inflammatory cytokine production, has translational implications deserving further analysis.
Collapse
Affiliation(s)
- Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Ivan Caiello
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Giulio Bontempi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Michela Terri
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Marta Tiberi
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- 15 REDINREN/RICORS2040, Madrid, Spain
| | - Alessandro Domenici
- Renal Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Paolo Menè
- Renal Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Giusi Prencipe
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- 15 REDINREN/RICORS2040, Madrid, Spain
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| |
Collapse
|
10
|
Moraru A, Dima ȘO, Tritean N, Oprița EI, Prelipcean AM, Trică B, Oancea A, Moraru I, Constantinescu-Aruxandei D, Oancea F. Bioactive-Loaded Hydrogels Based on Bacterial Nanocellulose, Chitosan, and Poloxamer for Rebalancing Vaginal Microbiota. Pharmaceuticals (Basel) 2023; 16:1671. [PMID: 38139798 PMCID: PMC10748236 DOI: 10.3390/ph16121671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Biocompatible drug-delivery systems for soft tissue applications are of high interest for the medical and pharmaceutical fields. The subject of this research is the development of hydrogels loaded with bioactive compounds (inulin, thyme essential oil, hydro-glycero-alcoholic extract of Vitis vinifera, Opuntia ficus-indica powder, lactic acid, citric acid) in order to support the vaginal microbiota homeostasis. The nanofibrillar phyto-hydrogel systems developed using the biocompatible polymers chitosan (CS), never-dried bacterial nanocellulose (NDBNC), and Poloxamer 407 (PX) incorporated the water-soluble bioactive components in the NDBNC hydrophilic fraction and the hydrophobic components in the hydrophobic core of the PX fraction. Two NDBNC-PX hydrogels and one NDBNC-PX-CS hydrogel were structurally and physical-chemically characterized using Fourier-transform infrared (FTIR) spectroscopy, X-ray diffraction (XRD), transmission electron microscopy (TEM), and rheology. The hydrogels were also evaluated in terms of thermo-responsive properties, mucoadhesion, biocompatibility, and prebiotic and antimicrobial effects. The mucin binding efficiency of hydrogel base systems was determined by the periodic acid/Schiff base (PAS) assay. Biocompatibility of hydrogel systems was determined by the MTT test using mouse fibroblasts. The prebiotic activity was determined using the probiotic strains Limosilactobacillus reuteri and Lactiplantibacillus plantarum subsp. plantarum. Antimicrobial activity was also assessed using relevant microbial strains, respectively, E. coli and C. albicans. TEM evidenced PX micelles of around 20 nm on NDBNC nanofibrils. The FTIR and XRD analyses revealed that the binary hydrogels are dominated by PX signals, and that the ternary hydrogel is dominated by CS, with additional particular fingerprints for the biocompounds and the hydrogel interaction with mucin. Rheology evidenced the gel transition temperatures of 18-22 °C for the binary hydrogels with thixotropic behavior and, respectively, no gel transition, with rheopectic behavior for the ternary hydrogel. The adhesion energies of the binary and ternary hydrogels were evaluated to be around 1.2 J/m2 and 9.1 J/m2, respectively. The hydrogels exhibited a high degree of biocompatibility, with the potential to support cell proliferation and also to promote the growth of lactobacilli. The hydrogel systems also presented significant antimicrobial and antibiofilm activity.
Collapse
Affiliation(s)
- Angela Moraru
- Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine Bucharest, Bd. Mărăști Nr. 59, Sector 1, 011464 Bucharest, Romania;
- S.C. Laboratoarele Medica Srl, Strada Frasinului Nr. 11, 075100 Otopeni, Romania;
| | - Ștefan-Ovidiu Dima
- Polymers and Bioresources Departments, National Institute for Research and Development in Chemistry and Petrochemistry—ICECHIM, Splaiul Independentei Nr. 202, Sector 6, 060021 Bucharest, Romania; (Ș.-O.D.); (N.T.); (B.T.)
| | - Naomi Tritean
- Polymers and Bioresources Departments, National Institute for Research and Development in Chemistry and Petrochemistry—ICECHIM, Splaiul Independentei Nr. 202, Sector 6, 060021 Bucharest, Romania; (Ș.-O.D.); (N.T.); (B.T.)
- Faculty of Biology, University of Bucharest, Splaiul Independentei Nr. 91-95, Sector 5, 050095 Bucharest, Romania
| | - Elena-Iulia Oprița
- Department of Cellular and Molecular Biology, National Institute of Research and Development for Biological Sciences, Splaiul Independentei Nr. 296, Sector 6, 060031 Bucharest, Romania; (E.-I.O.); (A.-M.P.); (A.O.)
| | - Ana-Maria Prelipcean
- Department of Cellular and Molecular Biology, National Institute of Research and Development for Biological Sciences, Splaiul Independentei Nr. 296, Sector 6, 060031 Bucharest, Romania; (E.-I.O.); (A.-M.P.); (A.O.)
| | - Bogdan Trică
- Polymers and Bioresources Departments, National Institute for Research and Development in Chemistry and Petrochemistry—ICECHIM, Splaiul Independentei Nr. 202, Sector 6, 060021 Bucharest, Romania; (Ș.-O.D.); (N.T.); (B.T.)
| | - Anca Oancea
- Department of Cellular and Molecular Biology, National Institute of Research and Development for Biological Sciences, Splaiul Independentei Nr. 296, Sector 6, 060031 Bucharest, Romania; (E.-I.O.); (A.-M.P.); (A.O.)
| | - Ionuț Moraru
- S.C. Laboratoarele Medica Srl, Strada Frasinului Nr. 11, 075100 Otopeni, Romania;
| | - Diana Constantinescu-Aruxandei
- Polymers and Bioresources Departments, National Institute for Research and Development in Chemistry and Petrochemistry—ICECHIM, Splaiul Independentei Nr. 202, Sector 6, 060021 Bucharest, Romania; (Ș.-O.D.); (N.T.); (B.T.)
| | - Florin Oancea
- Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine Bucharest, Bd. Mărăști Nr. 59, Sector 1, 011464 Bucharest, Romania;
- Polymers and Bioresources Departments, National Institute for Research and Development in Chemistry and Petrochemistry—ICECHIM, Splaiul Independentei Nr. 202, Sector 6, 060021 Bucharest, Romania; (Ș.-O.D.); (N.T.); (B.T.)
| |
Collapse
|
11
|
Chen R, Han X, Xu H, Xu J, Cao T, Shan Y, He F, Fang W, Li X. N-terminal domain of classical swine fever virus N pro induces proteasomal degradation of specificity protein 1 with reduced HDAC1 expression to evade from innate immune responses. J Virol 2023; 97:e0111523. [PMID: 37796122 PMCID: PMC10617410 DOI: 10.1128/jvi.01115-23] [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: 07/27/2023] [Accepted: 08/25/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE Of the flaviviruses, only CSFV and bovine viral diarrhea virus express Npro as the non-structural protein which is not essential for viral replication but functions to dampen host innate immunity. We have deciphered a novel mechanism with which CSFV uses to evade the host antiviral immunity by the N-terminal domain of its Npro to facilitate proteasomal degradation of Sp1 with subsequent reduction of HDAC1 and ISG15 expression. This is distinct from earlier findings involving Npro-mediated IRF3 degradation via the C-terminal domain. This study provides insights for further studies on how HDAC1 plays its role in antiviral immunity, and if and how other viral proteins, such as the core protein of CSFV, the nucleocapsid protein of porcine epidemic diarrhea virus, or even other coronaviruses, exert antiviral immune responses via the Sp1-HDAC1 axis. Such research may lead to a deeper understanding of viral immune evasion strategies as part of their pathogenetic mechanisms.
Collapse
Affiliation(s)
- Rong Chen
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Xiao Han
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Hankun Xu
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Jidong Xu
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Tong Cao
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Ying Shan
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Fang He
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Weihuan Fang
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| | - Xiaoliang Li
- Zhejiang University Institute of Preventive Veterinary Medicine & Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Akinsulie OC, Shahzad S, Ogunleye SC, Oladapo IP, Joshi M, Ugwu CE, Gbadegoye JO, Hassan FO, Adeleke R, Afolabi Akande Q, Adesola RO. Crosstalk between hypoxic cellular micro-environment and the immune system: a potential therapeutic target for infectious diseases. Front Immunol 2023; 14:1224102. [PMID: 37600803 PMCID: PMC10434535 DOI: 10.3389/fimmu.2023.1224102] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 08/22/2023] Open
Abstract
There are overwhelming reports on the promotional effect of hypoxia on the malignant behavior of various forms of cancer cells. This has been proposed and tested exhaustively in the light of cancer immunotherapy. However, there could be more interesting functions of a hypoxic cellular micro-environment than malignancy. There is a highly intricate crosstalk between hypoxia inducible factor (HIF), a transcriptional factor produced during hypoxia, and nuclear factor kappa B (NF-κB) which has been well characterized in various immune cell types. This important crosstalk shares common activating and inhibitory stimuli, regulators, and molecular targets. Impaired hydroxylase activity contributes to the activation of HIFs. Inflammatory ligands activate NF-κB activity, which leads to the expression of inflammatory and anti-apoptotic genes. The eventual sequelae of the interaction between these two molecular players in immune cells, either bolstering or abrogating functions, is largely cell-type dependent. Importantly, this holds promise for interesting therapeutic interventions against several infectious diseases, as some HIF agonists have helped prevent immune-related diseases. Hypoxia and inflammation are common features of infectious diseases. Here, we highlighted the role of this crosstalk in the light of functional immunity against infection and inflammation, with special focus on various innate and adaptive immune cells. Particularly, we discussed the bidirectional effects of this crosstalk in the regulation of immune responses by monocytes/macrophages, dendritic cells, neutrophils, B cells, and T cells. We believe an advanced understanding of the interplay between HIFs and NF-kB could reveal novel therapeutic targets for various infectious diseases with limited treatment options.
Collapse
Affiliation(s)
- Olalekan Chris Akinsulie
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Sammuel Shahzad
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Seto Charles Ogunleye
- College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | - Ifeoluwa Peace Oladapo
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Melina Joshi
- Center for Molecular Dynamics Nepal, Kathmandu, Nepal
| | - Charles Egede Ugwu
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
| | - Joy Olaoluwa Gbadegoye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Fasilat Oluwakemi Hassan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Richard Adeleke
- College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Qudus Afolabi Akande
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | | |
Collapse
|
13
|
Tseligka ED, Conzelmann S, Cambet Y, Schaer T, Negro F, Clément S. Identification of selective hepatitis delta virus ribozyme inhibitors by high-throughput screening of small molecule libraries. JHEP Rep 2022; 5:100652. [PMID: 36704052 PMCID: PMC9871325 DOI: 10.1016/j.jhepr.2022.100652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/24/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Background & Aims Chronic hepatitis delta is the most severe form of chronic viral hepatitis and is associated with faster progression towards cirrhosis, liver decompensation, and hepatocellular carcinoma. Hepatitis delta virus (HDV)'s tight dependency on hepatitis B virus and the host cell machinery for its life cycle limits the development of direct-acting antivirals. Thus, we aimed to identify compounds that could block HDV replication by targeting its antigenomic ribozyme. Methods We generated stable Huh7 human hepatoma cells expressing a reporter gene (Gaussia luciferase) either downstream (Gluc-2xRz) or upstream (2xRz-Gluc) of two HDV antigenomic ribozyme sequences. We performed high-throughput screening of three small molecule libraries. The secreted luciferase was measured as a readout of ribozyme inhibition upon addition of the molecules. Each plate was considered valid when the Z factor was >0.4. Specificity and toxicity evaluations were performed for the hits with a Z-score >5 and half-maximal inhibitory concentration was calculated by performing a dose-response experiment. Results A dose-dependent induction of luciferase expression was detected in Gluc-2xRz-transfected cells incubated with the antisense morpholino, suggesting that the catalytic activity of the ribozyme cloned downstream of the reporter gene was efficiently inhibited. Among the 6,644 compounds screened, we identified four compounds that showed a specific inhibitory effect on the HDV antigenomic ribozyme in Gluc-2xRz cells, i.e. three histone deacetylase inhibitors and the purine analogue 8-azaguanine. The latter also significantly decreased HDV replication (by 40%) in differentiated HepaRG cells six days post infection. Conclusion Using a novel cell culture model, we identified four small molecules active against the antigenomic HDV ribozyme. These results may provide insights into the structural requirements of molecules designed for the potent and specific inhibition of HDV replication. Impact and implications Chronic hepatitis delta is the most severe form of chronic viral hepatitis and is associated with faster progression towards cirrhosis, liver decompensation, and the development of hepatocellular carcinoma. Despite the current development of several new compounds, there is still a need for efficient antiviral treatments specifically targeting hepatitis delta virus (HDV). This work describes a novel cell culture model that allows for the high-throughput screening of compounds able to inhibit HDV ribozymes. We identified four small molecules active against the antigenomic HDV ribozyme (the ribozyme involved in the early step of HDV replication), with the strongest activity shown by 8-azaguanine, a purine analogue. Our data may provide insights into the structural requirements of molecules designed to inhibit HDV.
Collapse
Affiliation(s)
- Eirini D. Tseligka
- Department of Pathology and Immunology, University of Geneva, Switzerland
| | | | - Yves Cambet
- Department of Pathology and Immunology, University of Geneva, Switzerland,READS Unit, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Tifany Schaer
- Department of Pathology and Immunology, University of Geneva, Switzerland
| | - Francesco Negro
- Clinical Pathology, Geneva University Hospital, Geneva, Switzerland,Gastroenterology and Hepatology, Geneva University Hospital, Geneva, Switzerland,Corresponding author. Address: Clinical Pathology, and Gastroenterology and Hepatology, Geneva University Hospital, Geneva, Switzerland.
| | - Sophie Clément
- Department of Pathology and Immunology, University of Geneva, Switzerland
| |
Collapse
|
14
|
Bhat S, Rishi P, Chadha VD. Understanding the epigenetic mechanisms in SARS CoV-2 infection and potential therapeutic approaches. Virus Res 2022; 318:198853. [PMID: 35777502 PMCID: PMC9236910 DOI: 10.1016/j.virusres.2022.198853] [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: 02/16/2022] [Revised: 06/14/2022] [Accepted: 06/26/2022] [Indexed: 11/29/2022]
Abstract
COVID-19 pandemic caused by the Severe acute respiratory syndrome coronavirus 2 (SARS- CoV-2) has inflicted a global health challenge. Although the overwhelming escalation of mortality seen during the initial phase of the pandemic has reduced, emerging variants of SARS-CoV-2 continue to impact communities worldwide. Several studies have highlighted the association of gene specific epigenetic modifications in host cells with the pathogenesis and severity of the disease. Therefore, alongside the investigations into the virology and pathogenesis of SARS-CoV-2 infection, understanding the epigenetic mechanisms related to the disease is crucial for the rational design of effective targeted therapies. Here, we discuss the interaction of SARS-CoV-2 with the various epigenetic regulators and their subsequent contribution to the risk of disease severity and dysfunctional immune responses. Finally, we also highlight the use of epigenetically targeted drugs for the potential therapeutic interventions capable of eliminating viral infection and/or build effective immunity against it.
Collapse
Affiliation(s)
- Swati Bhat
- Center for Nuclear Medicine (U.I.E.A.S.T), South Campus, Panjab University, Sector 25, Chandigarh 160014, India.
| | - Praveen Rishi
- Department of Microbiology, South Campus, Panjab University, Sector 25, Chandigarh 160014, India.
| | - Vijayta D Chadha
- Center for Nuclear Medicine (U.I.E.A.S.T), South Campus, Panjab University, Sector 25, Chandigarh 160014, India.
| |
Collapse
|
15
|
Ta TM, Malik S, Anderson EM, Jones AD, Perchik J, Freylikh M, Sardo L, Klase ZA, Izumi T. Insights Into Persistent HIV-1 Infection and Functional Cure: Novel Capabilities and Strategies. Front Microbiol 2022; 13:862270. [PMID: 35572626 PMCID: PMC9093714 DOI: 10.3389/fmicb.2022.862270] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/21/2022] [Indexed: 12/23/2022] Open
Abstract
Although HIV-1 replication can be efficiently suppressed to undetectable levels in peripheral blood by combination antiretroviral therapy (cART), lifelong medication is still required in people living with HIV (PLWH). Life expectancies have been extended by cART, but age-related comorbidities have increased which are associated with heavy physiological and economic burdens on PLWH. The obstacle to a functional HIV cure can be ascribed to the formation of latent reservoir establishment at the time of acute infection that persists during cART. Recent studies suggest that some HIV reservoirs are established in the early acute stages of HIV infection within multiple immune cells that are gradually shaped by various host and viral mechanisms and may undergo clonal expansion. Early cART initiation has been shown to reduce the reservoir size in HIV-infected individuals. Memory CD4+ T cell subsets are regarded as the predominant cellular compartment of the HIV reservoir, but monocytes and derivative macrophages or dendritic cells also play a role in the persistent virus infection. HIV latency is regulated at multiple molecular levels in transcriptional and post-transcriptional processes. Epigenetic regulation of the proviral promoter can profoundly regulate the viral transcription. In addition, transcriptional elongation, RNA splicing, and nuclear export pathways are also involved in maintaining HIV latency. Although most proviruses contain large internal deletions, some defective proviruses may induce immune activation by expressing viral proteins or producing replication-defective viral-like particles. In this review article, we discuss the state of the art on mechanisms of virus persistence in the periphery and tissue and summarize interdisciplinary approaches toward a functional HIV cure, including novel capabilities and strategies to measure and eliminate the infected reservoirs and induce immune control.
Collapse
Affiliation(s)
- Tram M. Ta
- Department of Biological Sciences, Misher College of Arts and Sciences, University of the Sciences in Philadelphia, Philadelphia, PA, United States
| | - Sajjaf Malik
- Department of Biological Sciences, Misher College of Arts and Sciences, University of the Sciences in Philadelphia, Philadelphia, PA, United States
| | - Elizabeth M. Anderson
- Office of the Assistant Secretary for Health, Region 3, U.S. Department of Health and Human Services, Washington, DC, United States
| | - Amber D. Jones
- Department of Biological Sciences, Misher College of Arts and Sciences, University of the Sciences in Philadelphia, Philadelphia, PA, United States,Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jocelyn Perchik
- Department of Biological Sciences, Misher College of Arts and Sciences, University of the Sciences in Philadelphia, Philadelphia, PA, United States
| | - Maryann Freylikh
- Department of Biological Sciences, Misher College of Arts and Sciences, University of the Sciences in Philadelphia, Philadelphia, PA, United States
| | - Luca Sardo
- Department of Infectious Disease and Vaccines, Merck & Co., Inc., Kenilworth, NJ, United States
| | - Zackary A. Klase
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States,Center for Neuroimmunology and CNS Therapeutics, Institute of Molecular Medicine and Infectious Diseases, Drexel University of Medicine, Philadelphia, PA, United States
| | - Taisuke Izumi
- Department of Biological Sciences, Misher College of Arts and Sciences, University of the Sciences in Philadelphia, Philadelphia, PA, United States,*Correspondence: Taisuke Izumi,
| |
Collapse
|
16
|
Sixto-López Y, Correa-Basurto J. HDAC inhibition as neuroprotection in COVID-19 infection. Curr Top Med Chem 2022; 22:1369-1378. [PMID: 35240959 DOI: 10.2174/1568026622666220303113445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 11/22/2022]
Abstract
The SARS-CoV-2 virus is responsible of COVID-19 affecting millions of humans around the world. COVID-19 shows diverse clinical symptoms (fever, cough, fatigue, diarrhea, body aches, headaches, anosmia and hyposmia). Approximately 30% of the patients with COVID-19 showed neurological symptoms, these going from mild to severe manifestations including headache, dizziness, impaired consciousness, encephalopathy, anosmia, hypogeusia, hyposmia, psychology and psychiatry among others. The neurotropism of SARS-CoV-2 virus explains its neuroinvasion provoking neurological damage as acute demyelination, neuroinflammation etc. At molecular level, the COVID-19 patients had higher levels of cytokines and chemokines known as cytokines storms which disrupt the blood brain barrier allowing the entrance of monocytes and lymphocytes causing neuroinflammation, neurodegeneration and demyelination. In addition, ischemic, hemorrhagic strokes, seizures and encephalopathy have been observed due to the proinflammatory cytokines. In this sense, to avoid or decrease neurological damage due to SARS-CoV-2 infection, an early neuroprotective management should be adopted. Several approaches can be used; one of them includes the use of HDAC inhibitors (HDACi) due to their neuroprotective effects. Also, the HDACi down regulates the pro-inflammatory cytokines (IL-6 and TNF- decreasing the neurotoxicity. HDACi can also avoid and prevent the entrance of the virus into the Central nervous System (CNS) as well as decrease the virus replication by downregulating the virus receptors. Here we review the mechanisms that could explain how the SARS-CoV-2 virus could reach the CNS, induce the neurological damage and symptoms, as well as the possibility to use HDACi as neuroprotective therapy.
Collapse
Affiliation(s)
- Yudibeth Sixto-López
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, México. Plan de San Luis y Díaz Mirón S/N, Casco de Santo Tomas, Miguel Hidalgo, Mexico City, Mexico
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, México. Plan de San Luis y Díaz Mirón S/N, Casco de Santo Tomas, Miguel Hidalgo, Mexico City, Mexico
| |
Collapse
|
17
|
Quaas CE, Long DT. Targeting (de)acetylation: A Diversity of Mechanism and Disease. COMPREHENSIVE PHARMACOLOGY 2022:469-492. [DOI: 10.1016/b978-0-12-820472-6.00076-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
18
|
Lopes JR, Prokopczyk IM, Gerlack M, Man Chin C, Santos JLD. Design and Synthesis of Hybrid Compounds as Epigenetic Modifiers. Pharmaceuticals (Basel) 2021; 14:ph14121308. [PMID: 34959707 PMCID: PMC8709175 DOI: 10.3390/ph14121308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/22/2022] Open
Abstract
Epigenetic modifiers acting through polypharmacology mechanisms are promising compounds with which to treat several infectious diseases. Histone deacetylase (HDAC) enzymes, mainly class I, and extra-terminal bromodomains (BET) are involved in viral replication and the host response. In the present study, 10 compounds were designed, assisted by molecular docking, to act against HDAC class I and bromodomain-4 (BRD4). All the compounds were synthesized and characterized by analytical methods. Enzymatic assays were performed using HDAC-1, -4, and -11 and BRD4. Compounds (2-10) inhibited both HDAC class I, mainly HDAC-1 and -2, and reduced BRD4 activity. For HDAC-1, the inhibitory effect ranged from 8 to 95%, and for HDAC-2, these values ranged from 10 to 91%. Compounds (2-10) decreased the BRD4 activity by up to 25%. The multi-target effects of these compounds show desirable properties that could help to combat viral infections by acting through epigenetic mechanisms.
Collapse
|
19
|
Lu Y, Zhou X, Zhao W, Liao Z, Li B, Han P, Yang Y, Zhong X, Mo Y, Li P, Huang G, Xiao X, Zhang Z, Zhou X. Epigenetic Inactivation of Acetyl-CoA Acetyltransferase 1 Promotes the Proliferation and Metastasis in Nasopharyngeal Carcinoma by Blocking Ketogenesis. Front Oncol 2021; 11:667673. [PMID: 34485115 PMCID: PMC8415477 DOI: 10.3389/fonc.2021.667673] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/27/2021] [Indexed: 11/13/2022] Open
Abstract
The dysregulation of epigenetic modification and energy metabolism cooperatively contribute to the tumorigenesis of nasopharyngeal carcinoma (NPC). However, the detailed mechanisms underlying their joint contribution to NPC development and progression remain unclear. Here, we investigate the role of Acy1 Coenzyme A Acyltransferases1 (ACAT1), a key enzyme in the metabolic pathway of ketone bodies, in the proliferation and metastasis of NPC and to elucidate the underlying molecular mechanisms. Ketogenesis, plays a critical role in tumorigenesis. Previously, we reported two enzymes involved in ketone body metabolism mediate epigenetic silencing and act as tumor suppressor genes in NPC. Here, we identify another key enzyme, Acetyl-CoA acetyltransferase 1 (ACAT1), and show that its transcriptional inactivation in NPC is due to promoter hypermethylation. Ectopic overexpression of ACAT1 significantly suppressed the proliferation and colony formation of NPC cells in vitro. The migratory and invasive capacity of NPC cells was inhibited by ACAT1. The tumorigenesis of NPC cells overexpressing ACAT1 was decreased in vivo. Elevated ACAT1 in NPC cells was accompanied by an elevated expression of CDH1 and a reduced expression of vimentin and SPARC, strongly indicating that ACAT1 is involved in regulating epithelial-mesenchymal transition (EMT). We also found that ACAT1 contributes to increased intracellular levels of β-hydroxybutyrate (β-HB). Exogenously supplied β-HB significantly inhibits the growth of NPC cells in a dose-dependent manner. In summary, ACAT1 may function as a tumor suppressor via modulation of ketogenesis and could thus serve as a potential therapeutic target in NPC. In summary, our data suggest that regulation of ketogenesis may serve as adjuvant therapy in NPC.
Collapse
Affiliation(s)
- Yunliang Lu
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaohui Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Weilin Zhao
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhipeng Liao
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bo Li
- Department of Radiotherapy, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Peipei Han
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanping Yang
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xuemin Zhong
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yingxi Mo
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Ping Li
- Department of Pathology, Affiliated Stomatological Hospital of Guangxi Medical University, Nanning, China
| | - Guangwu Huang
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xue Xiao
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhe Zhang
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoying Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China
| |
Collapse
|
20
|
Bieszczad B, Garbicz D, Świtalska M, Dudek MK, Warszycki D, Wietrzyk J, Grzesiuk E, Mieczkowski A. Improved HDAC Inhibition, Stronger Cytotoxic Effect and Higher Selectivity against Leukemias and Lymphomas of Novel, Tricyclic Vorinostat Analogues. Pharmaceuticals (Basel) 2021; 14:851. [PMID: 34577551 PMCID: PMC8470702 DOI: 10.3390/ph14090851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 11/23/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors are a class of drugs used in the cancer treatment. Here, we developed a library of 19 analogues of Vorinostat, an HDAC inhibitor used in lymphomas treatment. In Vorinostat, we replaced the hydrophobic phenyl group with various tricyclic 'caps' possessing a central, eight-membered, heterocyclic ring, and investigated the HDAC activity and cytotoxic effect on the cancer and normal cell lines. We found that 3 out of the 19 compounds, based on dibenzo[b,f]azocin-6(5H)-one, 11,12-dihydrodibenzo[b,f]azocin-6(5H)-one, and benzo[b]naphtho[2,3-f][1,5]diazocine-6,14(5H,13H)-dione scaffolds, showed better HDACs inhibition than the referenced Vorinostat. In leukemic cell line MV4-11 and in the lymphoma cell line Daudi, three compounds showed lower IC50 values than Vorinostat. These compounds had higher activity and selectivity against MV4-11 and Daudi cell lines than reference Vorinostat. We also observed a strong correlation between HDACs inhibition and the cytotoxic effect. Cell lines derived from solid tumours: A549 (lung carcinoma) and MCF-7 (breast adenocarcinoma) as well as reference BALB/3T3 (normal murine fibroblasts) were less susceptible to compounds tested. Developed derivatives show improved properties than Vorinostat, thus they could be considered as possible agents for leukemia and lymphoma treatment.
Collapse
Affiliation(s)
- Bartosz Bieszczad
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland; (B.B.); (D.G.)
| | - Damian Garbicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland; (B.B.); (D.G.)
| | - Marta Świtalska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.Ś.); (J.W.)
| | - Marta K. Dudek
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland;
| | - Dawid Warszycki
- Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Cracow, Poland;
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland; (M.Ś.); (J.W.)
| | - Elżbieta Grzesiuk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland; (B.B.); (D.G.)
| | - Adam Mieczkowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland; (B.B.); (D.G.)
| |
Collapse
|
21
|
Targeting the latent human cytomegalovirus reservoir for T-cell-mediated killing with virus-specific nanobodies. Nat Commun 2021; 12:4436. [PMID: 34290252 PMCID: PMC8295288 DOI: 10.1038/s41467-021-24608-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/17/2021] [Indexed: 01/07/2023] Open
Abstract
Latent human cytomegalovirus (HCMV) infection is characterized by limited gene expression, making latent HCMV infections refractory to current treatments targeting viral replication. However, reactivation of latent HCMV in immunosuppressed solid organ and stem cell transplant patients often results in morbidity. Here, we report the killing of latently infected cells via a virus-specific nanobody (VUN100bv) that partially inhibits signaling of the viral receptor US28. VUN100bv reactivates immediate early gene expression in latently infected cells without inducing virus production. This allows recognition and killing of latently infected monocytes by autologous cytotoxic T lymphocytes from HCMV-seropositive individuals, which could serve as a therapy to reduce the HCMV latent reservoir of transplant patients.
Collapse
|
22
|
Pavel A, del Giudice G, Federico A, Di Lieto A, Kinaret PAS, Serra A, Greco D. Integrated network analysis reveals new genes suggesting COVID-19 chronic effects and treatment. Brief Bioinform 2021; 22:1430-1441. [PMID: 33569598 PMCID: PMC7929418 DOI: 10.1093/bib/bbaa417] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/13/2020] [Accepted: 12/19/2020] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 disease led to an unprecedented health emergency, still ongoing worldwide. Given the lack of a vaccine or a clear therapeutic strategy to counteract the infection as well as its secondary effects, there is currently a pressing need to generate new insights into the SARS-CoV-2 induced host response. Biomedical data can help to investigate new aspects of the COVID-19 pathogenesis, but source heterogeneity represents a major drawback and limitation. In this work, we applied data integration methods to develop a Unified Knowledge Space (UKS) and used it to identify a new set of genes associated with SARS-CoV-2 host response, both in vitro and in vivo. Functional analysis of these genes reveals possible long-term systemic effects of the infection, such as vascular remodelling and fibrosis. Finally, we identified a set of potentially relevant drugs targeting proteins involved in multiple steps of the host response to the virus.
Collapse
Affiliation(s)
- Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
| | - Giusy del Giudice
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
| | - Antonio Di Lieto
- Department of Forensic Psychiatry, Aarhus University, Aarhus, Denmark
| | - Pia A S Kinaret
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- BioMediTech Institute, Tampere University, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Aydemir MN, Aydemir HB, Korkmaz EM, Budak M, Cekin N, Pinarbasi E. Computationally predicted SARS-COV-2 encoded microRNAs target NFKB, JAK/STAT and TGFB signaling pathways. GENE REPORTS 2021; 22:101012. [PMID: 33398248 PMCID: PMC7773562 DOI: 10.1016/j.genrep.2020.101012] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/27/2020] [Accepted: 12/13/2020] [Indexed: 12/13/2022]
Abstract
Recently an outbreak that emerged in Wuhan, China in December 2019, spread to the whole world in a short time and killed >1,410,000 people. It was determined that a new type of beta coronavirus called severe acute respiratory disease coronavirus type 2 (SARS-CoV-2) was causative agent of this outbreak and the disease caused by the virus was named as coronavirus disease 19 (COVID19). Despite the information obtained from the viral genome structure, many aspects of the virus-host interactions during infection is still unknown. In this study we aimed to identify SARS-CoV-2 encoded microRNAs and their cellular targets. We applied a computational method to predict miRNAs encoded by SARS-CoV-2 along with their putative targets in humans. Targets of predicted miRNAs were clustered into groups based on their biological processes, molecular function, and cellular compartments using GO and PANTHER. By using KEGG pathway enrichment analysis top pathways were identified. Finally, we have constructed an integrative pathway network analysis with target genes. We identified 40 SARS-CoV-2 miRNAs and their regulated targets. Our analysis showed that targeted genes including NFKB1, NFKBIE, JAK1-2, STAT3-4, STAT5B, STAT6, SOCS1-6, IL2, IL8, IL10, IL17, TGFBR1-2, SMAD2-4, HDAC1-6 and JARID1A-C, JARID2 play important roles in NFKB, JAK/STAT and TGFB signaling pathways as well as cells' epigenetic regulation pathways. Our results may help to understand virus-host interaction and the role of viral miRNAs during SARS-CoV-2 infection. As there is no current drug and effective treatment available for COVID19, it may also help to develop new treatment strategies.
Collapse
Key Words
- ACE-2, angiotensin-converting enzyme 2
- AKT1, AKT serine/threonine kinase 1
- BCL2, BCL2 apoptosis regulator
- CDK1, cyclin dependent kinase 1
- CDKL2, cyclin dependent kinase like 2
- COVID19, new type corona virus disease
- CTNNB1, catenin beta 1
- CXCL1, C-X-C motif chemokine ligand 1
- CXCL10, C-X-C motif chemokine ligand 10
- CXCL11, C-X-C motif chemokine ligand 11
- CXCL16, C-X-C motif chemokine ligand 16
- CXCL9, C-X-C motif chemokine ligand 9
- E2F1, E2F transcription factor 1
- EIF4A1, eukaryotic translation initiation factor 4A1
- GRB2, growth factor receptor bound protein 2
- HDAC1, histone deacetylase 1
- HDAC2, histone deacetylase 2
- HDAC3, histone deacetylase 3
- HIF1A, hypoxia inducible factor 1 subunit alpha
- ICTV, International Committee on Taxonomy of Viruses
- IFNGR2, interferon gamma receptor 2
- IKBKE, inhibitor of nuclear factor kappa B kinase subunit epsilon
- IL10, interleukin 10
- IL13, interleukin 13
- IL15, interleukin 15
- IL16, interleukin 16
- IL17A, interleukin 17 A
- IL2, interleukin 2
- IL21, interleukin 21
- IL22, interleukin 22
- IL24, interleukin 24
- IL25, interleukin 25
- IL33, interleukin 33
- IL5, interleukin 5
- IL7, interleukin 7
- IL8, interleukin 8
- JAK/STAT
- JAK1, Janus kinase 1
- JAK2, Janus kinase 2
- JARID1A, lysine demethylase 5A
- JARID1B, lysine demethylase 5B
- JARID1C, lysine demethylase 5C
- JARID2, Jumonji and AT-rich interaction domain containing 2
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- MAPK1, mitogen-activated protein kinase 1
- MAPK3, mitogen-activated protein kinase 3
- MAPK4, mitogen-activated protein kinase 4
- MAPK6, mitogen-activated protein kinase 6
- MAPK7, mitogen-activated protein kinase 7
- NFKB
- NFKB1, nuclear factor kappa B subunit 1
- NFKBIE, NFKB inhibitor epsilon
- NOS3, nitric oxide synthase 3
- PANTHER, protein analysis through evolutionary relationships
- PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha
- PTEN, phosphatase and tensin homolog
- RB1, RB transcriptional corepressor 1
- RHOA, ras homolog family member A
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory disease coronavirus type 2
- SMAD2, SMAD family member 2
- SMAD3, SMAD family member 3
- SMAD4, SMAD family member 4
- SOCS1, suppressor of cytokine signaling 1
- SOCS3, suppressor of cytokine signaling 3
- SOCS4, suppressor of cytokine signaling 4
- SOCS5, suppressor of cytokine signaling 5
- SOCS6, suppressor of cytokine signaling 6
- SOS1, SOS Ras/Rac guanine nucleotide exchange factor 1
- SP1, Sp1 transcription factor
- STAT3, signal transducer and activator of transcription 3
- STAT4, signal transducer and activator of transcription 4
- STAT5B, signal transducer and activator of transcription 5B
- STAT6, signal transducer and activator of transcription 6
- SUMO1, small ubiquitin like modifier 1
- SUMO2, small ubiquitin like modifier 2
- TBP, TATA-box binding protein
- TGFB
- TGFBR1, transforming growth factor beta receptor 1
- TGFBR2, transforming growth factor beta receptor 2
- TMPRSS11A, transmembrane serine protease 11A
- TMPRSS4, transmembrane serine protease 4
- TNFRSF21, TNF receptor superfamily member 21
- WHO, World Health Organization
- miRNA
Collapse
Affiliation(s)
- Merve Nur Aydemir
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Habes Bilal Aydemir
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Gaziosmanpaşa University, Tokat, Turkey
| | - Ertan Mahir Korkmaz
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Mahir Budak
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Nilgun Cekin
- Sivas Cumhuriyet University, Faculty of Medicine, Department of Medical Biology, 58140 Sivas, Turkey
| | - Ergun Pinarbasi
- Sivas Cumhuriyet University, Faculty of Medicine, Department of Medical Biology, 58140 Sivas, Turkey
| |
Collapse
|
24
|
Khan MAAK, Islam ABMMK. SARS-CoV-2 Proteins Exploit Host's Genetic and Epigenetic Mediators for the Annexation of Key Host Signaling Pathways. Front Mol Biosci 2021; 7:598583. [PMID: 33585554 PMCID: PMC7872968 DOI: 10.3389/fmolb.2020.598583] [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: 08/25/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
The constant rise of the death toll and cases of COVID-19 has made this pandemic a serious threat to human civilization. Understanding of host-SARS-CoV-2 interaction in viral pathogenesis is still in its infancy. In this study, we utilized a blend of computational and knowledgebase approaches to model the putative virus-host interplay in host signaling pathways by integrating the experimentally validated host interactome proteins and differentially expressed host genes in SARS-CoV-2 infection. While searching for the pathways in which viral proteins interact with host proteins, we discovered various antiviral immune response pathways such as hypoxia-inducible factor 1 (HIF-1) signaling, autophagy, retinoic acid-inducible gene I (RIG-I) signaling, Toll-like receptor signaling, fatty acid oxidation/degradation, and IL-17 signaling. All these pathways can be either hijacked or suppressed by the viral proteins, leading to improved viral survival and life cycle. Aberration in pathways such as HIF-1 signaling and relaxin signaling in the lungs suggests the pathogenic lung pathophysiology in COVID-19. From enrichment analysis, it was evident that the deregulated genes in SARS-CoV-2 infection might also be involved in heart development, kidney development, and AGE-RAGE signaling pathway in diabetic complications. Anomalies in these pathways might suggest the increased vulnerability of COVID-19 patients with comorbidities. Moreover, we noticed several presumed infection-induced differentially expressed transcription factors and epigenetic factors, such as miRNAs and several histone modifiers, which can modulate different immune signaling pathways, helping both host and virus. Our modeling suggests that SARS-CoV-2 integrates its proteins in different immune signaling pathways and other cellular signaling pathways for developing efficient immune evasion mechanisms while leading the host to a more complicated disease condition. Our findings would help in designing more targeted therapeutic interventions against SARS-CoV-2.
Collapse
|
25
|
Abstract
Acetylation was initially discovered as a post-translational modification (PTM) on the unstructured, highly basic N-terminal tails of eukaryotic histones in the 1960s. Histone acetylation constitutes part of the "histone code", which regulates chromosome compaction and various DNA processes such as gene expression, recombination, and DNA replication. In bacteria, nucleoid-associated proteins (NAPs) are responsible these functions in that they organize and compact the chromosome and regulate some DNA processes. The highly conserved DNABII family of proteins are considered functional homologues of eukaryotic histones despite having no sequence or structural conservation. Within the past decade, a growing interest in Nε-lysine acetylation led to the discovery that hundreds of bacterial proteins are acetylated with diverse cellular functions, in direct contrast to the original thought that this was a rare phenomenon. Similarly, other previously undiscovered bacterial PTMs, like serine, threonine, and tyrosine phosphorylation, have also been characterized. In this review, the various PTMs that were discovered among DNABII family proteins, specifically histone-like protein (HU) orthologues, from large-scale proteomic studies are discussed. The functional significance of these modifications and the enzymes involved are also addressed. The discovery of novel PTMs on these proteins begs this question: is there a histone-like code in bacteria?
Collapse
Affiliation(s)
- Valerie J Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, New Jersey 08103, United States
| |
Collapse
|
26
|
Management of epigenomic networks entailed in coronavirus infections and COVID-19. Clin Epigenetics 2020; 12:118. [PMID: 32758273 PMCID: PMC7404079 DOI: 10.1186/s13148-020-00912-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
Coronaviruses (CoVs) are highly diverse single-stranded RNA viruses owing to their susceptibility to numerous genomic mutations and recombination. Such viruses involve human and animal pathogens including the etiologic agents of acute respiratory tract illnesses: severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and the highly morbific SARS-CoV-2. Coronavirus disease 2019 (COVID-19), an emerging disease with a quick rise in infected cases and deaths, was recently identified causing a worldwide pandemic. COVID-19 disease outcomes were found to increase in elderly and patients with a compromised immune system. Evidences indicated that the main culprit behind COVID-19 deaths is the cytokine storm, which is illustrated by an uncontrolled over-production of soluble markers of inflammation. The regulation process of coronavirus pathogenesis through molecular mechanism comprise virus-host interactions linked to viral entry, replication and transcription, escape, and immune system control. Recognizing coronavirus infections and COVID-19 through epigenetics lens will lead to potential alteration in gene expression thus limiting coronavirus infections. Focusing on epigenetic therapies reaching clinical trials, clinically approved epigenetic-targeted agents, and combination therapy of antivirals and epigenetic drugs is currently considered an effective and valuable approach for viral replication and inflammatory overdrive control.
Collapse
|
27
|
Pasquereau S, Totoson P, Nehme Z, Abbas W, Kumar A, Verhoeven F, Prati C, Wendling D, Demougeot C, Herbein G. Impact of glucocorticoids on systemic sirtuin 1 expression and activity in rats with adjuvant-induced arthritis. Epigenetics 2020; 16:132-143. [PMID: 32615849 DOI: 10.1080/15592294.2020.1790789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The class III histone deacetylase sirtuin 1 (SIRT1) plays a pivotal role in numerous biological and physiological functions, including inflammation. An association between SIRT1 and proinflammatory cytokines might exist. In addition to their important role in inflammation associated with rheumatoid arthritis (RA), proinflammatory cytokines mediate the development of systemic effects. Here, we evaluated systemic SIRT1 expression and enzymatic activity, in peripheral blood mononuclear cells (PBMCs) and in liver isolated from rats with adjuvant-induced arthritis (AIA), treated or not with low or high doses of glucocorticoids (GCs). We also measured the production of tumour necrosis factor alpha (TNF) and interleukin-1 beta (IL-1 beta) in PBMCs and liver. We found that SIRT1 expression and activity increased in PBMCs of AIA rats compared to healthy controls and decreased under GC treatment. Similarly, we observed an increased SIRT1 activity in the liver of AIA rats compared to healthy controls which decreased under high doses of GCs. We also found an increase in IL-1 beta and TNF levels in the liver of AIA rats compared to healthy controls, which decreased under high doses of GC. We did not observe a significant correlation between SIRT1 activity and proinflammatory cytokine production in PBMC or liver. In contrast, a strong positive correlation was found between the liver levels of TNF and IL-1 beta (rho=0.9503, p=7.5x10-21). Our results indicate that increased inflammation in AIA rats compared to healthy control is accompanied by an increased SIRT1 activity in both PBMCs and liver, which could be decreased under GC treatment.
Collapse
Affiliation(s)
- Sébastien Pasquereau
- Pathogens & Inflammation/EPILAB Laboratory, UPRES EA 4266, SFR FED 4234, University of Franche-Comté, Université Bourgogne Franche-Comté (UBFC) , Besançon, France
| | - Perle Totoson
- Pepite EA4267, Université Bourgogne Franche-Comté (UBFC) , Besançon, France
| | - Zeina Nehme
- Pathogens & Inflammation/EPILAB Laboratory, UPRES EA 4266, SFR FED 4234, University of Franche-Comté, Université Bourgogne Franche-Comté (UBFC) , Besançon, France
| | - Wasim Abbas
- Pathogens & Inflammation/EPILAB Laboratory, UPRES EA 4266, SFR FED 4234, University of Franche-Comté, Université Bourgogne Franche-Comté (UBFC) , Besançon, France
| | - Amit Kumar
- Pathogens & Inflammation/EPILAB Laboratory, UPRES EA 4266, SFR FED 4234, University of Franche-Comté, Université Bourgogne Franche-Comté (UBFC) , Besançon, France
| | - Frank Verhoeven
- Pepite EA4267, Université Bourgogne Franche-Comté (UBFC) , Besançon, France.,Department of Rheumatology, CHRU Besançon , Besançon, France
| | - Clément Prati
- Pepite EA4267, Université Bourgogne Franche-Comté (UBFC) , Besançon, France.,Department of Rheumatology, CHRU Besançon , Besançon, France
| | - Daniel Wendling
- Pathogens & Inflammation/EPILAB Laboratory, UPRES EA 4266, SFR FED 4234, University of Franche-Comté, Université Bourgogne Franche-Comté (UBFC) , Besançon, France.,Department of Rheumatology, CHRU Besançon , Besançon, France
| | - Céline Demougeot
- Pepite EA4267, Université Bourgogne Franche-Comté (UBFC) , Besançon, France
| | - Georges Herbein
- Pathogens & Inflammation/EPILAB Laboratory, UPRES EA 4266, SFR FED 4234, University of Franche-Comté, Université Bourgogne Franche-Comté (UBFC) , Besançon, France.,Department of Virology, CHRU Besançon , Besançon, France
| |
Collapse
|
28
|
Al-Eitan LN, Alghamdi MA, Tarkhan AH, Al-Qarqaz FA. Genome-wide identification of methylated CpG sites in nongenital cutaneous warts. BMC Med Genomics 2020; 13:100. [PMID: 32641122 PMCID: PMC7346436 DOI: 10.1186/s12920-020-00745-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 06/19/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Low-risk HPV infection has not been the subject of epigenetic investigation. The present study was carried out in order to investigate the methylation status of CpG sites in non-genital cutaneous warts. METHODS Genomic DNA was extracted from 24 paired epidermal samples of warts and normal skin. DNA samples were bisulfite converted and underwent genome-wide methylation profiling using the Infinium MethylationEPIC BeadChip Kit. RESULTS From a total of 844,234 CpG sites, 56,960 and 43,040 CpG sites were found to be hypo- and hypermethylated, respectively, in non-genital cutaneous warts. The most differentially methylated CpG sites in warts were located within the C10orf26, FAM83H-AS1, ZNF644, LINC00702, GSAP, STAT5A, HDAC4, NCALD, and EXOC4 genes. CONCLUSION Non-genital cutaneous warts exhibit a unique CpG methylation signature.
Collapse
Affiliation(s)
- Laith N Al-Eitan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan.
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Mansour A Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
| | - Amneh H Tarkhan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Firas A Al-Qarqaz
- Department of Internal Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
- Division of Dermatology, Department of Internal Medicine, King Abdullah University Hospital Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
29
|
Kuroda S, Miyagawa Y, Sato Y, Yamamoto M, Adachi K, Kinoh H, Goins WF, Cohen JB, Glorioso JC, Taniai N, Yoshida H, Okada T. Protocol Optimization for the Production of the Non-Cytotoxic JΔNI5 HSV Vector Deficient in Expression of Immediately Early Genes. Mol Ther Methods Clin Dev 2020; 17:612-621. [PMID: 32300608 PMCID: PMC7150431 DOI: 10.1016/j.omtm.2020.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/12/2020] [Indexed: 12/15/2022]
Abstract
Non-toxic herpes simplex virus (HSV) vectors can be generated by functional deletion of all immediate-early (IE) genes, providing a benign vehicle with potential for gene therapy. However, deletion of multiple IE genes raises manufacturing concerns and thus limits clinical application of these vectors. To address this issue, we previously developed a novel production cell line, called U2OS-ICP4/27, by lentiviral transduction of human osteosarcoma U2OS cells with two essential HSV IE genes, ICP4 and ICP27. To optimize the process of vector manufacturing on this platform, we evaluated several cell culture parameters of U2OS-ICP4/27 for high-titer and -quality production of non-toxic HSV vectors, revealing that the yields and functionality of these vectors can be significantly influenced by culturing conditions. We also found that several chemical compounds can enhance the replication of non-toxic HSV vectors and their release from producer cells into the supernatants. Notably, the vector produced by our optimized protocol displayed a greatly improved vector yield and quality and showed elevated transgene expression in cultures of primary dorsal root ganglion neurons. Taken together, our optimized production approach emerges as a relevant protocol for high-yield and high-quality preparation of non-toxic HSV-based gene therapy vectors.
Collapse
Affiliation(s)
- Seiji Kuroda
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
- Department of Surgery, Nippon Medical School Musashikosugi Hospital, Kawasaki, Japan
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Yoshitaka Miyagawa
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Yuriko Sato
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Motoko Yamamoto
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Kumi Adachi
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Hiromi Kinoh
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - William F. Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Justus B. Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Joseph C. Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Nobuhiko Taniai
- Department of Surgery, Nippon Medical School Musashikosugi Hospital, Kawasaki, Japan
| | - Hiroshi Yoshida
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
30
|
SIRT1 enhances hepatitis virus B transcription independent of hepatic autophagy. Biochem Biophys Res Commun 2020; 527:64-70. [PMID: 32446392 DOI: 10.1016/j.bbrc.2020.04.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 02/02/2023]
Abstract
Autophagy is an intracellular process that can lead to the degradation of malfunctioned proteins and damaged organelles to maintain homeostasis during cellular stress. Here, we evaluated the change in hepatitis B virus (HBV) production by regulating hepatic autophagy in HBV-producing cells. We examined focusing on a relation with a positive autophagy regulator, sirtuin1 (SIRT1). Starvation and rapamycin treatment induced autophagy with increasing SIRT1 protein, HBc protein and pregenomic RNA (pgRNA) levels in HBV- producing cells. Knockdown of Atg7 or Atg13 suppressed hepatic autophagy, and it did not change SIRT1 protein, HBc protein or pgRNA levels in HBV- producing cells. Resveratrol, which increases SIRT1 expression and activity, promoted autophagy and increased HBc protein and pgRNA levels. siRNA-mediated knockdown of SIRT1 inhibited autophagy and decreased HBc protein and pgRNA levels. In SIRT1-knockdown cells, starvation promoted autophagy but did not increase HBc protein and pgRNA levels. In conclusion, HBc protein and pgRNA levels are upregulated not by the autophagic process itself but by the SIRT1 expression level.
Collapse
|
31
|
Herbein G, Nehme Z. Tumor Control by Cytomegalovirus: A Door Open for Oncolytic Virotherapy? MOLECULAR THERAPY-ONCOLYTICS 2020; 17:1-8. [PMID: 32300639 PMCID: PMC7150429 DOI: 10.1016/j.omto.2020.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Belonging to the herpesviridae family, human cytomegalovirus (HCMV) is a well-known ubiquitous pathogen that establishes a lifelong infection in humans. Recently, a beneficial tumor-cytoreductive role of CMV infection has been defined in human and animal models. Described as a potential anti-tumoral activity, HCMV modulates the tumor microenvironment mainly by inducing cell death through apoptosis and prompting a robust stimulatory effect on the immune cells infiltrating the tumor tissue. However, major current limitations embrace transient protective effect and a viral dissemination potential in immunosuppressed hosts. The latter could be counteracted through direct viral intratumoral delivery, use of non-human strains, or even defective CMV vectors to ascertain transformed cells-selective tropism. This potential oncolytic activity could be complemented by tackling further platforms, namely combination with immune checkpoint inhibitors or epigenetic therapy, as well as the use of second-generation chimeric oncovirus, for instance HCMV/HSV-1 oncolytic virus. Overall, preliminary data support the use of CMV in viral oncolytic therapy as a viable option, establishing thus a potential new modality, where further assessment through extensive basic research armed by molecular biotechnology is compulsory.
Collapse
Affiliation(s)
- Georges Herbein
- Department Pathogens & Inflammation-EPILAB, UPRES EA4266, University of Franche-Comté, University of Bourgogne Franche-Comté, 25030 Besançon, France.,Department of Virology, CHRU Besancon, 25030 Besançon, France
| | - Zeina Nehme
- Department Pathogens & Inflammation-EPILAB, UPRES EA4266, University of Franche-Comté, University of Bourgogne Franche-Comté, 25030 Besançon, France.,Université Libanaise 1003, Beirut, Lebanon
| |
Collapse
|
32
|
Nehme Z, Pasquereau S, Herbein G. Targeting histone epigenetics to control viral infections. HISTONE MODIFICATIONS IN THERAPY 2020. [PMCID: PMC7453269 DOI: 10.1016/b978-0-12-816422-8.00011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During the past decades, many studies have significantly broadened our understanding of complex virus-host interactions to control chromatin structure and dynamics.1, 2 However, the role and impact of such modifications during viral infections is not fully revealed. Indeed, this type of regulation is bidirectional between the virus and the host. While viral replication and gene expression are significantly impacted by histone modifications on the viral chromatin,3 studies have shown that some viral pathogens dynamically manipulate cellular epigenetic factors to enhance their own survival and pathogenesis, as well as escape the immune system defense lines.4 In this dynamic, histone posttranslational modifications (PTMs) appear to play fundamental roles in the regulation of chromatin structure and recruitment of other factors.5 Genuinely, those PTMs play a vital role in lytic infection, latency reinforcement, or, conversely, viral reactivation.6 In this chapter, we will examine and review the involvement of histone modifications as well as their potential manipulation to control infections during various viral life cycle stages, highlighting their prospective implications in the clinical management of human immunodeficiency virus (HIV), herpes simplex virus (HSV), human cytomegalovirus (HCMV), hepatitis B and C viruses (HBV and HCV, respectively), Epstein–Barr virus (EBV), and other viral diseases. Targeting histone modifications is critical in setting the treatment of chronic viral infections with both lytic and latent stages (HIV, HCMV, HSV, RSV), virus-induced cancers (HBV, HCV, EBV, KSHV, HPV), and epidemic/emerging viruses (e.g. influenza virus, arboviruses).
Collapse
|
33
|
Ran X, Ao Z, Olukitibi T, Yao X. Characterization of the Role of Host Cellular Factor Histone Deacetylase 10 during HIV-1 Replication. Viruses 2019; 12:v12010028. [PMID: 31888084 PMCID: PMC7020091 DOI: 10.3390/v12010028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 12/28/2022] Open
Abstract
To date, a series of histone deacetylases have been documented to restrict HIV-1 replication at different steps. In this study, we identified histone deacetylase 10 (HDAC10) as an inhibitory factor against HIV-1 replication. Our results showed that endogenous HDAC10 is downregulated at the transcriptional level during HIV-1 replication. By knocking down HDAC10 in CD4+ T cells with specific shRNAs, we observed that the downregulation of HDAC10 significantly facilitates viral replication. Moreover, RQ-PCR analysis revealed that the downregulation of HDAC10 increased viral integrated DNA. Further, we identified that HDAC10 interacts with the HIV-1 integrase (IN) and that the region of residues from 55 to 165 in the catalytic domain of IN is required for HDAC10 binding. Interestingly, we found that the interaction between HDAC10 and IN specifically decreases the interaction between IN and cellular protein lens epithelium-derived growth factor (LEDGF/p75), which consequently leads to the inhibition of viral integration. In addition, we have investigated the role of HDAC10 in the late stage of viral replication by detecting the infectiousness of progeny virus produced from HDAC10 knockdown cells or HDAC10 overexpressing cells and revealed that the progeny virus infectivity is increased in the HDAC10 downregulated cells, but decreased in the HDAC10 overexpressed cells. Overall, these findings provide evidence that HDAC10 acts as a cellular inhibitory factor at the early and late stages of HIV-1 replication.
Collapse
|
34
|
Simões M, Freitas FB, Leitão A, Martins C, Ferreira F. African swine fever virus replication events and cell nucleus: New insights and perspectives. Virus Res 2019; 270:197667. [PMID: 31319112 DOI: 10.1016/j.virusres.2019.197667] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/13/2019] [Accepted: 07/14/2019] [Indexed: 12/30/2022]
Abstract
African swine fever (ASF) is currently matter for major concerns in global swine industry as it is highly contagious and causes acute fatal haemorrhagic fever in domestic pigs and wild boar. The absence of effective vaccines and treatments pushes ASF control to relay on strict sanitary and stamping out measures with costly socio-economic impacts. The current epidemic scenario of fast spreading throughout Asiatic countries impels further studies on prevention and combat strategies against ASF. Herein we review knowledge on African Swine Fever Virus (ASFV) interactions with the host cell nucleus and on the functional properties of different viral DNA-replication related proteins. This entails, the confirmation of an intranuclear viral DNA replication phase, the characterization of cellular DNA damage responses (DDR), the subnuclear compartments disruption due to viral modulation, and the unravelling of the biological role of several viral proteins (A104R, I215 L, P1192R, QP509 L and Q706 L), so to contribute to underpin rational strategies for vaccine candidates development.
Collapse
Affiliation(s)
- Margarida Simões
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal; Laboratório de Virologia, Instituto Nacional de Investigação Agrária e Veterinária (INIAV), Quinta do Marquês, 2780-157, Oeiras, Portugal
| | - Ferdinando B Freitas
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Alexandre Leitão
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Carlos Martins
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Fernando Ferreira
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal.
| |
Collapse
|
35
|
Chen W, Yi C, Jin L. The Role of Nicotinamide Adenine Dinucleotide in the Pathogenesis of Rheumatoid Arthritis: Potential Implications for Treatment. EUROPEAN MEDICAL JOURNAL 2018. [DOI: 10.33590/emj/10312205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic, inflammatory, autoimmune disease characterised by small joint swelling, deformity, and dysfunction. Its exact aetiology is unclear. Current treatment approaches do not control harmful autoimmune attacks or prevent irreversible damage without considerable side effects. Nicotinamide adenine dinucleotide (NAD+), an important hydrogen carrier in mitochondrial respiration and oxidative phosphorylation, is the major determinant of redox state in the cell. NAD+ metabolites act as degradation substrates for a wide range of enzymes, such as sirtuins, poly-ADP-ribose polymerases, ADP-ribosyltransferases, and CD38. The roles of NAD+ have expanded beyond its role as a coenzyme, linking cellular metabolism to inflammation signalling and immune response. The aim of this review is to illustrate the role of NAD+-related enzymes in the pathogenesis of RA and highlight the potential therapeutic role of NAD+ in RA.
Collapse
Affiliation(s)
- Weiqian Chen
- Department of Rheumatology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Caihong Yi
- Department of Rheumatology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Jin
- Department of Rheumatology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
36
|
Kozlov MV, Malikova AZ, Kamarova KA, Konduktorov KA, Kochetkov SN. Synthesis of Pyridyl-4-Oxy-Substituted N-Hydroxy Amides of Cinnamic Acid as New Inhibitors of Histone Deacetylase Activity and Hepatitis C Virus Replication. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2018. [DOI: 10.1134/s1068162018040118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
37
|
The Process and Strategy for Developing Selective Histone Deacetylase 3 Inhibitors. Molecules 2018; 23:molecules23030551. [PMID: 29498635 PMCID: PMC6017514 DOI: 10.3390/molecules23030551] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/21/2018] [Accepted: 02/27/2018] [Indexed: 12/31/2022] Open
Abstract
Histone deacetylases (HDACs) are epigenetic drug targets that have gained major scientific attention. Inhibition of these important regulatory enzymes is used to treat cancer, and has the potential to treat a host of other diseases. However, currently marketed HDAC inhibitors lack selectivity for the various HDAC isoenzymes. Several studies have shown that HDAC3, in particular, plays an important role in inflammation and degenerative neurological diseases, but the development of selective HDAC3 inhibitors has been challenging. This review provides an up-to-date overview of selective HDAC3 inhibitors, and aims to support the development of novel HDAC3 inhibitors in the future.
Collapse
|
38
|
Guo H, Zhang J, Wang Y, Bu C, Zhou Y, Fang Q. Comparative Proteomic Analysis of Lysine Acetylation in Fish CIK Cells Infected with Aquareovirus. Int J Mol Sci 2017; 18:E2419. [PMID: 29135940 PMCID: PMC5713387 DOI: 10.3390/ijms18112419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/10/2017] [Accepted: 11/12/2017] [Indexed: 01/06/2023] Open
Abstract
Grass carp (Ctenopharyngodon idellus) is an important worldwide commercial freshwater culture species. However, grass carp reovirus (GCRV) causes serious hemorrhagic disease in fingerlings and yearlings of fishes. To understand the molecular pathogenesis of host cells during GCRV infection, intensive proteomic quantification analysis of lysine acetylation in Ctenopharyngodon idella kidney (CIK) cells was performed. Using dimethylation labeling-based quantitative proteomics, 832 acetylated proteins with 1391 lysine acetylation sites were identified in response to GCRV infection, among which 792 proteins with 1323 sites were quantifiable. Bioinformatics analysis showed that differentially expressed lysine acetylated proteins are involved in diverse cellular processes and associated with multifarious functions, suggesting that extensive intracellular activities were changed upon viral infection. In addition, extensive alterations on host-protein interactions at the lysine acetylation level were also detected. Further biological experiments showed that the histone deacetylases (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) could significantly suppress the GCRV replication. To our knowledge, this is the first to reveal the proteome-wide changes in host cell acetylome with aquatic virus infection. The results provided in this study laid a basis for further understanding the host response to aquareovirus infection in the post-translational modification aspect by regulating cell lysine acetylation conducive to viral replication.
Collapse
Affiliation(s)
- Hong Guo
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Jie Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Yaping Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Chen Bu
- Jingjie PTM BioLab (Hangzhou) Co., Ltd., Hangzhou 310018, China.
| | - Yanyan Zhou
- Jingjie PTM BioLab (Hangzhou) Co., Ltd., Hangzhou 310018, China.
| | - Qin Fang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
39
|
Sodium phenylbutyrate abrogates African swine fever virus replication by disrupting the virus-induced hypoacetylation status of histone H3K9/K14. Virus Res 2017; 242:24-29. [DOI: 10.1016/j.virusres.2017.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/06/2017] [Accepted: 09/11/2017] [Indexed: 02/08/2023]
|
40
|
Abstract
Nε-Lysine acetylation is now recognized as an abundant posttranslational modification (PTM) that influences many essential biological pathways. Advancements in mass spectrometry-based proteomics have led to the discovery that bacteria contain hundreds of acetylated proteins, contrary to the prior notion of acetylation events being rare in bacteria. Although the mechanisms that regulate protein acetylation are still not fully defined, it is understood that this modification is finely tuned via both enzymatic and nonenzymatic mechanisms. The opposing actions of Gcn5-related N-acetyltransferases (GNATs) and deacetylases, including sirtuins, provide the enzymatic control of lysine acetylation. A nonenzymatic mechanism of acetylation has also been demonstrated and proven to be prominent in bacteria, as well as in mitochondria. The functional consequences of the vast majority of the identified acetylation sites remain unknown. From studies in mammalian systems, acetylation of critical lysine residues was shown to impact protein function by altering its structure, subcellular localization, and interactions. It is becoming apparent that the same diversity of functions can be found in bacteria. Here, we review current knowledge of the mechanisms and the functional consequences of acetylation in bacteria. Additionally, we discuss the methods available for detecting acetylation sites, including quantitative mass spectrometry-based methods, which promise to promote this field of research. We conclude with possible future directions and broader implications of the study of protein acetylation in bacteria.
Collapse
|
41
|
Nagesh PT, Hussain M, Galvin HD, Husain M. Histone Deacetylase 2 Is a Component of Influenza A Virus-Induced Host Antiviral Response. Front Microbiol 2017; 8:1315. [PMID: 28769891 PMCID: PMC5511851 DOI: 10.3389/fmicb.2017.01315] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 06/29/2017] [Indexed: 11/13/2022] Open
Abstract
Host cells produce variety of antiviral factors that create an antiviral state and target various stages of influenza A virus (IAV) life cycle to inhibit infection. However, IAV has evolved various strategies to antagonize those antiviral factors. Recently, we reported that a member of class I host histone deacetylases (HDACs), HDAC1 possesses an anti-IAV function. Herein, we provide evidence that HDAC2, another class I member and closely related to HDAC1 in structure and function, also possesses anti-IAV properties. In turn, IAV, like HDAC1, dysregulates HDAC2, mainly at the polypeptide level through proteasomal degradation to potentially minimize its antiviral effect. We found that IAV downregulated the HDAC2 polypeptide level in A549 cells in an H1N1 strain-independent manner by up to 47%, which was recovered to almost 100% level in the presence of proteasome-inhibitor MG132. A further knockdown in HDAC2 expression by up to 90% via RNA interference augmented the growth kinetics of IAV in A549 cells by more than four-fold after 24 h of infection. Furthermore, the knockdown of HDAC2 expression decreased the IAV-induced phosphorylation of the transcription factor, Signal Transducer and Activator of Transcription I (STAT1) and the expression of interferon-stimulated gene, viperin in infected cells by 41 and 53%, respectively. The role of HDAC2 in viperin expression was analogous to that of HDAC1, but it was not in the phosphorylation of STAT1. This indicated that, like HDAC1, HDAC2 is a component of IAV-induced host innate antiviral response and performs both redundant and non-redundant functions vis-a-vis HDAC1; however, IAV dysregulates them both in a redundant manner.
Collapse
Affiliation(s)
- Prashanth T Nagesh
- Department of Microbiology and Immunology, University of OtagoDunedin, New Zealand.,Department of Microbiology, New York University School of Medicine, New YorkNY, United States
| | - Mazhar Hussain
- Department of Microbiology and Immunology, University of OtagoDunedin, New Zealand
| | - Henry D Galvin
- Department of Microbiology and Immunology, University of OtagoDunedin, New Zealand
| | - Matloob Husain
- Department of Microbiology and Immunology, University of OtagoDunedin, New Zealand
| |
Collapse
|
42
|
Structural insights of SmKDAC8 inhibitors: Targeting Schistosoma epigenetics through a combined structure-based 3D QSAR, in vitro and synthesis strategy. Bioorg Med Chem 2017; 25:2105-2132. [DOI: 10.1016/j.bmc.2017.02.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 11/24/2022]
|
43
|
Romani B, Kamali Jamil R, Hamidi-Fard M, Rahimi P, Momen SB, Aghasadeghi MR, Allahbakhshi E. HIV-1 Vpr reactivates latent HIV-1 provirus by inducing depletion of class I HDACs on chromatin. Sci Rep 2016; 6:31924. [PMID: 27550312 PMCID: PMC4994036 DOI: 10.1038/srep31924] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/29/2016] [Indexed: 12/25/2022] Open
Abstract
HIV-1 Vpr is an accessory protein that induces proteasomal degradation of multiple proteins. We recently showed that Vpr targets class I HDACs on chromatin for proteasomal degradation. Here we show that Vpr induces degradation of HDAC1 and HDAC3 in HIV-1 latently infected J-Lat cells. Degradation of HDAC1 and HDAC3 was also observed on the HIV-1 LTR and as a result, markers of active transcription were recruited to the viral promoter and induced viral activation. Knockdown of HDAC1 and HDAC3 activated the latent HIV-1 provirus and complementation with HDAC3 inhibited Vpr-induced HIV-1 reactivation. Viral reactivation and degradation of HDAC1 and HDAC3 was conserved among Vpr proteins of HV-1 group M. Serum Vpr isolated from patients or the release of virion-incorporated Vpr from viral lysates also activated HIV-1 in latently infected cell lines and PBMCs from HIV-1 infected patients. Our results indicate that Vpr counteracts HIV-1 latency by inducing proteasomal degradation of HDAC1 and 3 leading to reactivation of the viral promoter.
Collapse
Affiliation(s)
- Bizhan Romani
- Cellular and Molecular Research Center (CMRC), Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, 61357-15794, Iran.,Department of Biology, Faculty of Science, University of Isfahan, Isfahan, 81746-73441, Iran
| | - Razieh Kamali Jamil
- Department of Human Viral Vaccines, Razi Vaccine and Serum Research Institute, Karaj, 31976-19751, Iran
| | - Mojtaba Hamidi-Fard
- Hepatitis and AIDS Department, Pasteur Institute of Iran, Tehran, 13169-43551, Iran
| | - Pooneh Rahimi
- Hepatitis and AIDS Department, Pasteur Institute of Iran, Tehran, 13169-43551, Iran
| | - Seyed Bahman Momen
- Pilot Biotechnology Department, Pasteur Institute of Iran, Tehran, 13169-43551, Iran
| | | | - Elham Allahbakhshi
- Cellular and Molecular Research Center (CMRC), Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, 61357-15794, Iran
| |
Collapse
|
44
|
Influenza A Virus Dysregulates Host Histone Deacetylase 1 That Inhibits Viral Infection in Lung Epithelial Cells. J Virol 2016; 90:4614-4625. [PMID: 26912629 DOI: 10.1128/jvi.00126-16] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/16/2016] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED Viruses dysregulate the host factors that inhibit virus infection. Here, we demonstrate that human enzyme, histone deacetylase 1 (HDAC1) is a new class of host factor that inhibits influenza A virus (IAV) infection, and IAV dysregulates HDAC1 to efficiently replicate in epithelial cells. A time-dependent decrease in HDAC1 polypeptide level was observed in IAV-infected cells, reducing to <50% by 24 h of infection. A further depletion (97%) of HDAC1 expression by RNA interference increased the IAV growth kinetics, increasing it by >3-fold by 24 h and by >6-fold by 48 h of infection. Conversely, overexpression of HDAC1 decreased the IAV infection by >2-fold. Likewise, a time-dependent decrease in HDAC1 activity, albeit with slightly different kinetics to HDAC1 polypeptide reduction, was observed in infected cells. Nevertheless, a further inhibition of deacetylase activity increased IAV infection in a dose-dependent manner. HDAC1 is an important host deacetylase and, in addition to its role as a transcription repressor, HDAC1 has been lately described as a coactivator of type I interferon response. Consistent with this property, we found that inhibition of deacetylase activity either decreased or abolished the phosphorylation of signal transducer and activator of transcription I (STAT1) and expression of interferon-stimulated genes, IFITM3, ISG15, and viperin in IAV-infected cells. Furthermore, the knockdown of HDAC1 expression in infected cells decreased viperin expression by 58% and, conversely, the overexpression of HDAC1 increased it by 55%, indicating that HDAC1 is a component of IAV-induced host type I interferon antiviral response. IMPORTANCE Influenza A virus (IAV) continues to significantly impact global public health by causing regular seasonal epidemics, occasional pandemics, and zoonotic outbreaks. IAV is among the successful human viral pathogens that has evolved various strategies to evade host defenses, prevent the development of a universal vaccine, and acquire antiviral drug resistance. A comprehensive knowledge of IAV-host interactions is needed to develop a novel and alternative anti-IAV strategy. Host produces a variety of factors that are able to fight IAV infection by employing various mechanisms. However, the full repertoire of anti-IAV host factors and their antiviral mechanisms has yet to be identified. We have identified here a new host factor, histone deacetylase 1 (HDAC1) that inhibits IAV infection. We demonstrate that HDAC1 is a component of host innate antiviral response against IAV, and IAV undermines HDAC1 to limit its role in antiviral response.
Collapse
|
45
|
Simões M, Rino J, Pinheiro I, Martins C, Ferreira F. Alterations of Nuclear Architecture and Epigenetic Signatures during African Swine Fever Virus Infection. Viruses 2015; 7:4978-96. [PMID: 26389938 PMCID: PMC4584302 DOI: 10.3390/v7092858] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 12/11/2022] Open
Abstract
Viral interactions with host nucleus have been thoroughly studied, clarifying molecular mechanisms and providing new antiviral targets. Considering that African swine fever virus (ASFV) intranuclear phase of infection is poorly understood, viral interplay with subnuclear domains and chromatin architecture were addressed. Nuclear speckles, Cajal bodies, and promyelocytic leukaemia nuclear bodies (PML-NBs) were evaluated by immunofluorescence microscopy and Western blot. Further, efficient PML protein knockdown by shRNA lentiviral transduction was used to determine PML-NBs relevance during infection. Nuclear distribution of different histone H3 methylation marks at lysine’s 9, 27 and 36, heterochromatin protein 1 isoforms (HP1α, HPβ and HPγ) and several histone deacetylases (HDACs) were also evaluated to assess chromatin status of the host. Our results reveal morphological disruption of all studied subnuclear domains and severe reduction of viral progeny in PML-knockdown cells. ASFV promotes H3K9me3 and HP1β foci formation from early infection, followed by HP1α and HDAC2 nuclear enrichment, suggesting heterochromatinization of host genome. Finally, closeness between DNA damage response factors, disrupted PML-NBs, and virus-induced heterochromatic regions were identified. In sum, our results demonstrate that ASFV orchestrates spatio-temporal nuclear rearrangements, changing subnuclear domains, relocating Ataxia Telangiectasia Mutated Rad-3 related (ATR)-related factors and promoting heterochromatinization, probably controlling transcription, repressing host gene expression, and favouring viral replication.
Collapse
Affiliation(s)
- Margarida Simões
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - José Rino
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Inês Pinheiro
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.
| | - Carlos Martins
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Fernando Ferreira
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida Universidade Técnica, 1300-477 Lisboa, Portugal.
| |
Collapse
|
46
|
Wendling D, Abbas W, Godfrin-Valnet M, Kumar A, Guillot X, Khan KA, Vidon C, Coquard L, Toussirot E, Prati C, Herbein G. Dysregulated serum IL-23 and SIRT1 activity in peripheral blood mononuclear cells of patients with rheumatoid arthritis. PLoS One 2015; 10:e0119981. [PMID: 25799392 PMCID: PMC4370395 DOI: 10.1371/journal.pone.0119981] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 01/16/2015] [Indexed: 12/24/2022] Open
Abstract
Sirtuin 1 (Sirt1) is a class III histone deacetylase (HDAC) that modulates gene expression and is involved in the regulation of proinflammatory cytokines. Interleukin-23 (IL-23) is produced by activated macrophages and dendritic cells and could fuel the progression of rheumatoid arthritis (RA). The goal of our study was to evaluate serum IL-23 levels and both Sirt1 activity and expression in peripheral blood mononuclear cells (PBMCs) in patients with RA compared to healthy controls (HC) and to determine the relationship between Sirt1 activity/expression and IL-23 levels. We assessed apoptosis in PBMCs of RA patients and its association with Sirt1 expression and serum IL-23. Serum IL-23 levels were increased in RA patients in comparison with controls. We found a positive correlation between the levels of serum IL-23 and serum IL-6 in RA patients. Decreased cytoplasmic Sirt1 activity was observed in RA patients with severe disease compared to HC. The expression of Sirt1 protein was significantly decreased in PBMCs of RA patients compared to HC using western blotting. Serum IL-23 levels correlated positively with the cytoplasmic Sirt1 activity in RA patients. Apoptosis rate of PBMCs isolated from RA patients was increased compared to HC and correlated negatively with the expression of Sirt1 protein and serum IL-23 levels. Levels of serum IL-23 and Sirt1 activity and expression were disturbed in RA parallel to increased PBMC apoptosis. Our findings might provide the rationale for the development of new therapeutic approaches in RA.
Collapse
Affiliation(s)
- Daniel Wendling
- Department of Rheumatology, Centre Hospitalier Régional Universitaire, Besançon, France
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| | - Wasim Abbas
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| | - Marie Godfrin-Valnet
- Department of Rheumatology, Centre Hospitalier Régional Universitaire, Besançon, France
| | - Amit Kumar
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| | - Xavier Guillot
- Department of Rheumatology, Centre Hospitalier Régional Universitaire, Besançon, France
| | - Kashif Aziz Khan
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| | - Claire Vidon
- Department of Rheumatology, Centre Hospitalier Régional Universitaire, Besançon, France
| | - Laurie Coquard
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| | - Eric Toussirot
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
- Clinical Investigation Biotherapy Center506, Centre Hospitalier Régional Universitaire, Besançon, France
- Department of Therapeutics, University of Franche Comté, Besançon, France
| | - Clément Prati
- Department of Rheumatology, Centre Hospitalier Régional Universitaire, Besançon, France
| | - Georges Herbein
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
- Department of Virology, Centre Hospitalier Régional Universitaire, Besançon, France
- Structure Fédérative de Recherche 4234, University of Franche-Comté, Besançon, France
- * E-mail:
| |
Collapse
|
47
|
Transcriptome responses of the host Trichoplusia ni to infection by the baculovirus Autographa californica multiple nucleopolyhedrovirus. J Virol 2014; 88:13781-97. [PMID: 25231311 DOI: 10.1128/jvi.02243-14] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Productive infection of Trichoplusia ni cells by the baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV) leads to expression of ~156 viral genes and results in dramatic cell remodeling. How the cell transcriptome responds to viral infection was unknown due to the lack of a reference genome and transcriptome for T. ni. We used an ~60-Gb RNA sequencing (RNA-seq) data set from infected and uninfected T. ni cells to generate and annotate a de novo transcriptome assembly of approximately 70,322 T. ni unigenes (assembled transcripts), representing the 48-h infection cycle. Using differential gene expression analysis, we found that the majority of host transcripts were downregulated after 6 h postinfection (p.i.) and throughout the remainder of the infection. In contrast, 5.7% (4,028) of the T. ni unigenes were upregulated during the early period (0 to 6 h p.i.), followed by a decrease through the remainder of the infection cycle. Also, a small subset of genes related to metabolism and stress response showed a significant elevation of transcript levels at 18 and 24 h p.i. but a decrease thereafter. We also examined the responses of genes belonging to a number of specific pathways of interest, including stress responses, apoptosis, immunity, and protein trafficking. We identified specific pathway members that were upregulated during the early phase of the infection. Combined with the parallel analysis of AcMNPV expression, these results provide both a broad and a detailed view of how baculovirus infection impacts the host cell transcriptome to evade cellular defensive responses, to modify cellular biosynthetic pathways, and to remodel cell structure. IMPORTANCE Baculoviruses are insect-specific DNA viruses that are highly pathogenic to their insect hosts. In addition to their use for biological control of certain insects, baculoviruses also serve as viral vectors for numerous biotechnological applications, such as mammalian cell transduction and protein expression for vaccine production. While there is considerable information regarding viral gene expression in infected cells, little is known regarding responses of the host cell to baculovirus infection. In these studies, we assembled a cell transcriptome from the host Trichoplusia ni and used that transcriptome to analyze changes in host cell gene expression throughout the infection cycle. The study was performed in parallel with a prior study of changes in viral gene expression. Combined, these studies provide an unprecedented new level of detail and an overview of events in the infection cycle, and they will stimulate new experimental approaches to understand, modify, and utilize baculoviruses for a variety of applications.
Collapse
|
48
|
Godfrin-Valnet M, Khan KA, Guillot X, Prati C, Baud L, Abbas W, Toussirot E, Wendling D, Herbein G. Sirtuin 1 activity in peripheral blood mononuclear cells of patients with osteoporosis. Med Sci Monit Basic Res 2014; 20:142-5. [PMID: 25228047 PMCID: PMC4181306 DOI: 10.12659/msmbr.891372] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background Sirtuin 1 (SIRT1) is a class III histone deacetylase that may play a critical role in several biological functions, including lifespan, stress, and inflammation. Our main objective was to evaluate SIRT1 activity in peripheral blood mononuclear cells (PBMCs) in patients with osteoporosis and to analyze the relationship between the SIRT 1 activity and markers of inflammation and bone remodelling. Material/Methods We performed a prospective monocentric study of patients with osteoporosis and measured the nuclear and cytoplasmic activities of SIRT1 in PBMCs. Levels of proinflammatory cytokines were assessed in culture supernatants of PBMCs isolated from the osteoporosis patients. The level of serum C-terminal cross-linking telopeptide of type I collagen (CTX), a marker of bone resorption, was measured in the serum of osteoporosis patients. Results Sixteen women with osteoporosis were included. A statistically significant correlation between the cytoplasmic and nuclear SIRT 1 activities was found in PBMCs of patients with osteoporosis. Although non-significant, we observed a negative trend between nuclear SIRT 1 activity and the rate of serum CTX and a positive trend between IL-6 and CTX levels in patients with osteoporosis. Conclusions This study shows that the cytoplasmic and nuclear SIRT 1 activities are measurable in circulating PBMCs of patients with osteoporosis and that these 2 activities are correlated. The potential role of inflammation in bone resorption in patients with osteoporosis was also studied.
Collapse
Affiliation(s)
- Marie Godfrin-Valnet
- Department of Rheumatology, Centre Hospitalier Régional Universitaire (CHRU) Besançon, Besançon, France
| | - Kashif A Khan
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| | - Xavier Guillot
- Department of Rheumatology, Centre Hospitalier Régional Universitaire (CHRU) Besançon, Besançon, France
| | - Clément Prati
- Department of Rheumatology, Centre Hospitalier Régional Universitaire (CHRU) Besançon, Besançon, France
| | - Lucile Baud
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| | - Wasim Abbas
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| | - Eric Toussirot
- Department of Rheumatology, Centre Hospitalier Régional Universitaire (CHRU) Besançon, Besançon, France
| | - Daniel Wendling
- Department of Rheumatology, Centre Hospitalier Régional Universitaire (CHRU) Besançon, Besançon, France
| | - Georges Herbein
- Pathogens & Inflammation Laboratory, University of Franche-Comté, Besançon, France
| |
Collapse
|
49
|
Utilization of Boron Compounds for the Modification of Suberoyl Anilide Hydroxamic Acid as Inhibitor of Histone Deacetylase Class II Homo sapiens. Adv Bioinformatics 2014; 2014:104823. [PMID: 25214833 PMCID: PMC4158260 DOI: 10.1155/2014/104823] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/12/2014] [Accepted: 08/12/2014] [Indexed: 01/26/2023] Open
Abstract
Histone deacetylase (HDAC) has a critical function in regulating gene expression. The inhibition of HDAC has developed as an interesting anticancer research area that targets biological processes such as cell cycle, apoptosis, and cell differentiation. In this study, an HDAC inhibitor that is available commercially, suberoyl anilide hydroxamic acid (SAHA), has been modified to improve its efficacy and reduce the side effects of the compound. Hydrophobic cap and zinc-binding group of these compounds were substituted with boron-based compounds, whereas the linker region was substituted with p-aminobenzoic acid. The molecular docking analysis resulted in 8 ligands with ΔGbinding value more negative than the standards, SAHA and trichostatin A (TSA). That ligands were analyzed based on the nature of QSAR, pharmacological properties, and ADME-Tox. It is conducted to obtain a potent inhibitor of HDAC class II Homo sapiens. The screening process result gave one best ligand, Nova2 (513246-99-6), which was then further studied by molecular dynamics simulations.
Collapse
|
50
|
Wendling D, Vidon C, Abbas W, Guillot X, Toussirot E, Herbein G. Sirt1 activity in peripheral blood mononuclear cells from patients with rheumatoid arthritis. Joint Bone Spine 2014; 81:462-3. [PMID: 24746477 DOI: 10.1016/j.jbspin.2014.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 02/13/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Daniel Wendling
- Department of Rheumatology, CHRU de Besançon, boulevard Fleming, 25030 Besançon, France; University of Franche-Comté, UPRES EA 4266 Pathogens & Inflammation Laboratory, SFR FED4234, Besançon, France.
| | - Claire Vidon
- Department of Rheumatology, CHRU de Besançon, boulevard Fleming, 25030 Besançon, France
| | - Wasim Abbas
- University of Franche-Comté, UPRES EA 4266 Pathogens & Inflammation Laboratory, SFR FED4234, Besançon, France
| | - Xavier Guillot
- Department of Rheumatology, CHRU de Besançon, boulevard Fleming, 25030 Besançon, France
| | - Eric Toussirot
- Department of Rheumatology, CHRU de Besançon, boulevard Fleming, 25030 Besançon, France; University of Franche-Comté, UPRES EA 4266 Pathogens & Inflammation Laboratory, SFR FED4234, Besançon, France
| | - Georges Herbein
- University of Franche-Comté, UPRES EA 4266 Pathogens & Inflammation Laboratory, SFR FED4234, Besançon, France; Department of Virology, CHRU de Besançon, place Saint-Jacques, 25030 Besançon, France.
| |
Collapse
|