1
|
Wagih N, Abdel-Rahman IM, El-Koussi NA, El-Din A Abuo-Rahma G. Anticancer benzimidazole derivatives as inhibitors of epigenetic targets: a review article. RSC Adv 2025; 15:966-1010. [PMID: 39807197 PMCID: PMC11726184 DOI: 10.1039/d4ra05014b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality worldwide. One of the primary causes of cancer development and progression is epigenetic dysregulation, which is a heritable modification that alters gene expression without changing the DNA sequence. Therefore, targeting these epigenetic changes has emerged as a promising therapeutic strategy. Benzimidazole derivatives have gained attention for their potent epigenetic modulatory effects as they interact with various epigenetic targets, including DNA methyltransferases, histone deacetylases and histone methyltransferases. This review provides a comprehensive overview of benzimidazole derivatives that inhibit different acetylation and methylation reader, writer and eraser epigenetic targets. Herein, we emphasize the therapeutic potential of these compounds in developing targeted, less toxic cancer therapies. Presently, some promising benzimidazole derivatives have entered clinical trials and shown great advancements in the fields of hematological and solid malignancy therapies. Accordingly, we highlight the recent advancements in benzimidazole research as epigenetic agents that could pave the way for designing new multi-target drugs to overcome resistance and improve clinical outcomes for cancer patients. This review can help researchers in designing new anticancer benzimidazole derivatives with better properties.
Collapse
Affiliation(s)
- Nardin Wagih
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University New Minia 61111 Egypt
| | - Islam M Abdel-Rahman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University New Minia 61111 Egypt
| | - Nawal A El-Koussi
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University New Minia 61111 Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, Assiut University 71526 Assiut Egypt
| | - Gamal El-Din A Abuo-Rahma
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University 61519 Minia Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University New Minia 61111 Egypt
| |
Collapse
|
2
|
Pang Z, Tang A, He Y, Fan J, Yang Q, Tong Y, Fan H. Neurological complications caused by SARS-CoV-2. Clin Microbiol Rev 2024; 37:e0013124. [PMID: 39291997 PMCID: PMC11629622 DOI: 10.1128/cmr.00131-24] [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] [Indexed: 09/19/2024] Open
Abstract
SUMMARYSARS-CoV-2 can not only cause respiratory symptoms but also lead to neurological complications. Research has shown that more than 30% of SARS-CoV-2 patients present neurologic symptoms during COVID-19 (A. Pezzini and A. Padovani, Nat Rev Neurol 16:636-644, 2020, https://doi.org/10.1038/s41582-020-0398-3). Increasing evidence suggests that SARS-CoV-2 can invade both the central nervous system (CNS) (M.S. Xydakis, M.W. Albers, E.H. Holbrook, et al. Lancet Neurol 20: 753-761, 2021 https://doi.org/10.1016/S1474-4422(21)00182-4 ) and the peripheral nervous system (PNS) (M.N. Soares, M. Eggelbusch, E. Naddaf, et al. J Cachexia Sarcopenia Muscle 13:11-22, 2022, https://doi.org/10.1002/jcsm.12896), resulting in a variety of neurological disorders. This review summarized the CNS complications caused by SARS-CoV-2 infection, including encephalopathy, neurodegenerative diseases, and delirium. Additionally, some PNS disorders such as skeletal muscle damage and inflammation, anosmia, smell or taste impairment, myasthenia gravis, Guillain-Barré syndrome, ICU-acquired weakness, and post-acute sequelae of COVID-19 were described. Furthermore, the mechanisms underlying SARS-CoV-2-induced neurological disorders were also discussed, including entering the brain through retrograde neuronal or hematogenous routes, disrupting the normal function of the CNS through cytokine storms, inducing cerebral ischemia or hypoxia, thus leading to neurological complications. Moreover, an overview of long-COVID-19 symptoms is provided, along with some recommendations for care and therapeutic approaches of COVID-19 patients experiencing neurological complications.
Collapse
Affiliation(s)
- Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ao Tang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yujie He
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Junfen Fan
- Department of Neurology, Institute of Cerebrovascular Diseases Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qingmao Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huahao Fan
- School of Life Sciences, Tianjin University, Tianjin, China
| |
Collapse
|
3
|
Zhang SY, Zhang LY, Wen R, Yang N, Zhang TN. Histone deacetylases and their inhibitors in inflammatory diseases. Biomed Pharmacother 2024; 179:117295. [PMID: 39146765 DOI: 10.1016/j.biopha.2024.117295] [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: 06/19/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Despite considerable research efforts, inflammatory diseases remain a heavy burden on human health, causing significant economic losses annually. Histone deacetylases (HDACs) play a significant role in regulating inflammation (via histone and non-histone protein deacetylation) and chromatin structure and gene expression regulation. Herein, we present a detailed description of the different HDACs and their functions and analyze the role of HDACs in inflammatory diseases, including pro-inflammatory cytokine production reduction, immune cell function modulation, and anti-inflammatory cell activity enhancement. Although HDAC inhibitors have shown broad inflammatory disease treatment potentials, their clinical applicability remains limited because of their non-specific effects, adverse effects, and drug resistance. With further research and insight, these inhibitors are expected to become important tools for the treatment of a wide range of inflammatory diseases. This review aims to explore the mechanisms and application prospects of HDACs and their inhibitors in multiple inflammatory diseases.
Collapse
Affiliation(s)
- Sen-Yu Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Li-Ying Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Tie-Ning Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
4
|
Moreno-Pérez O, Merino E, Manuel Ramos J, Carlos Rodríguez J, Diaz C, Mas P, Reus S, Sánchez-Martínez R, Boix V, Chico-Sánchez P, Sánchez-Payá J, Portilla J. Valproic acid could help in the fight against COVID-19: A case-control study. Neurologia 2024; 39:549-554. [PMID: 39232592 DOI: 10.1016/j.nrleng.2023.12.009] [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: 11/12/2021] [Accepted: 01/22/2022] [Indexed: 09/06/2024] Open
Abstract
OBJECTIVE There is early evidence about Valproic acid (VPA) antiviral effect. Our aim was to investigate the incidence and severity of SARS-CoV-2 infection in VPA users as compared with the general population. MATERIAL AND METHODS A case-control study nested within a cohort, carried out between March 1 and December 17, 2020. Retrospectively, we identified confirmed SARS-CoV-2 infection patients exposed to VPA in our health department (defined as case). We ascertained VPA regimen (all the time (AT) (292 days) or at least 20% of the study period (notAT) (≥58 days) and if VPA levels were in therapeutic range (ATR) (50-100mcg/mL) in the last 24 months. We calculated the cumulative incidence of SARS-CoV-2 infection and hospital admission in the cases, comparing it with the general unexposed VPA population (controls). RESULTS During the study period, 6183 PCR+ were detected among 281,035 inhabitants, of these, 746 were hospitalized. 691 patients were on VPA notAT and 628 (90.1%) AT. The indication for VPA use was epilepsy in 54.9%. The incidence of PCR+ was 1.736% (OR 0.785 (95%CI 0.443-1.390) and 1.910% (OR 0.865 (95%CI 0.488-1.533), on VPA notAT and VPA AT patients, respectively vs. 2.201% in people without VPA regimen. Those patients with VPA ATR had a lower risk of PCR + (OR 0.233 (95%CI 0.057-0.951) notAT; OR 0.218 (95%CI 0.053-0.890) AT). Hospital admission incidence was lower in patient on VPA (OR was 0.543 (95% CI 0.076-3.871). CONCLUSION Patients with VPA within the therapeutic range had a reduction of SARS-Cov-2 infection incidence greater than 75%. There is a downward trend in the risk of COVID-19 admission by SARS-CoV-2 in patients on VPA therapy. These findings warrant further investigation.
Collapse
Affiliation(s)
- O Moreno-Pérez
- Endocrinology and Nutrition department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain; Clinical Medicine Department, Miguel Hernández University, Elche, Spain
| | - E Merino
- Unit of Infectious Diseases, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain.
| | - J Manuel Ramos
- Internal Medicine Department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain; Clinical Medicine Department, Miguel Hernández University, Elche, Spain
| | - J Carlos Rodríguez
- Microbiology Department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain; Miguel Hernández University, Elche, Spain
| | - C Diaz
- Neurology Department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain; Clinical Medicine Department, Miguel Hernández University, Elche, Spain
| | - P Mas
- Pharmacy Department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - S Reus
- Unit of Infectious Diseases, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain; Clinical Medicine Department, Miguel Hernández University, Elche, Spain
| | - R Sánchez-Martínez
- Internal Medicine department, Alicante General University Hospital - Alicante Institute of Sanitary and Biomedical Research (ISABIAL), Alicante, Spain
| | - V Boix
- Unit of Infectious Diseases, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain; Clinical Medicine Department, Miguel Hernández University, Elche, Spain
| | - P Chico-Sánchez
- Preventive Department, Alicante General University Hospital - Alicante Institute of Sanitary and Biomedical Research (ISABIAL), Alicante, Spain
| | - J Sánchez-Payá
- Preventive Department, Alicante General University Hospital - Alicante Institute of Sanitary and Biomedical Research (ISABIAL), Alicante, Spain
| | - J Portilla
- Internal Medicine Department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain; Clinical Medicine Department, Miguel Hernández University, Elche, Spain
| |
Collapse
|
5
|
Watson A, Shah P, Lee D, Liang S, Joshi G, Metitiri E, Chowdhury WH, Bacich D, Dube P, Xiang Y, Hanley D, Martinez-Sobrido L, Rodriguez R. Valproic acid use is associated with diminished risk of contracting COVID-19, and diminished disease severity: Epidemiologic and in vitro analysis reveal mechanistic insights. PLoS One 2024; 19:e0307154. [PMID: 39093886 PMCID: PMC11296636 DOI: 10.1371/journal.pone.0307154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
The SARS-CoV-2 pandemic has caused unprecedented worldwide infections from persistent mutant variants with various degrees of infectivity and virulence. The elusiveness of a highly penetrant, worldwide vaccination strategy suggests that the complete eradication of SARS-CoV-2 is unlikely. Even with the advent of new antiviral agents, the disease burden worldwide continues to exceed current preventative and therapeutic strategies. Greater interest has been placed towards the development of affordable,broadly effective antiviral therapeutics. Here, we report that the small branched-chain fatty acid Valproic acid (VPA), approved for maintenance of seizure and bipolar disorder, has a novel anti- coronavirus activity that can be augmented with the addition of a long-chain, polyunsaturated omega-3 fatty acid, Docosahexaenoic acid (DHA). An EMR-based epidemiological study of patients tested for COVID-19 demonstrated a correlation exists between a reduced infection rate in patients treated withVPA of up to 25%, as well as a decreased risk of emergency room visits, hospitalization, ICU admission,and use of mechanical ventilation. In vitro studies have demonstrated that VPA modifies gene expression in MRC5 cells. Interestingly, VPA correlates with the inhibition of several SARS-CoV2 interacting genes and the greater inhibition of alpha-coronavirus HCoV-229E (a "common cold" virus) and SARS-CoV2. The VPA-DHA combination activates pre-existing intracellular antiviral mechanisms normally repressed by coronaviruses. Gene expression profiles demonstrate subtle differences in overall gene expression between VPA-treated and VPA-DHA-treated cells. HCoV-229E infection caused an intensely different response with a marked induction of multiple intracellular inflammatory genes. Changes in gene expression took at least 24 hours to manifest and most likely why prior drug screens failed to identify any antiviral VPA activity despite in silico predictions. This report demonstrates an interaction between HDAC inhibition and the potent activation of cellular antiviral responses. A foundation now exists for a low-cost, highly effective antiviral strategy when supplemented with DHA.
Collapse
Affiliation(s)
- Amanda Watson
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Pankil Shah
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Doug Lee
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Sitai Liang
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Geeta Joshi
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Ediri Metitiri
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Wasim H. Chowdhury
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Dean Bacich
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Peter Dube
- Boehringer Ingelheim in Ames, Ames, Iowa, United States of America
| | - Yan Xiang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio San Antonio, Texas, United States of America
| | - Daniel Hanley
- Department of Neurology & Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | | | - Ronald Rodriguez
- Department of Medical Education, and Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
6
|
Alzahrani KJ. Repurposing of Anti-Cancer Drugs Against Moderate and Severe COVID Infection: A Network-Based Systems Biological Approach. Niger J Clin Pract 2024; 27:950-957. [PMID: 39212430 DOI: 10.4103/njcp.njcp_873_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The COVID-19 pandemic caused by SARS-CoV-2 is an unparalleled health risk, needing fast antiviral medication development. One of the most effective strategies for developing therapies against novel and emerging viruses is drug repurposing. Recently, systems biology approaches toward the discovery of repurposing medications are gaining prominence. AIM This study aimed to implement a systems biology approach to identify crucial drug targets as well as potential drug candidates against COVID infection. METHODS Our approach utilizes differential gene expression in COVID conditions that enable the construction of a protein-protein interaction (PPI) network. Core clusters were extracted from this network, followed by molecular enrichment analysis. This process identified critical drug targets and potential drug candidates targeting various stages of COVID-19 infection. RESULTS The network was built using the top 200 differently expressed genes in mild, moderate, and severe COVID-19 infections. Top 3 clusters for each disease condition were identified, representing the core mechanism of the network. Molecular enrichment revealed the majority of the pathways in the mild state were associated with transcription regulation, protein folding, angiogenesis, and cytokine-signaling pathways. Whereas, the enriched pathways in moderate and severe disease states were predominately linked with the immune system and apoptotic processes, which include NF-kappaB signaling, cytokine signaling, TNF-mediated signaling, and oxidative stress-induced cell death. Further analysis identifies 28 potential drugs that can be repurposed to treat moderate and severe COVID-19, most of which are currently used in cancer treatment. CONCLUSION Interestingly, some of the proposed drugs have demonstrated inhibitory effects against SARS-CoV-2, as supported by literature evidence. Overall, the drug repurposing method described here will help develop potential antiviral medications to treat emerging COVID strains.
Collapse
Affiliation(s)
- K J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| |
Collapse
|
7
|
Zhou C, Zhao D, Wu C, Wu Z, Zhang W, Chen S, Zhao X, Wu S. Role of histone deacetylase inhibitors in non-neoplastic diseases. Heliyon 2024; 10:e33997. [PMID: 39071622 PMCID: PMC11283006 DOI: 10.1016/j.heliyon.2024.e33997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Background Epigenetic dysregulation has been implicated in the development and progression of a variety of human diseases, but epigenetic changes are reversible, and epigenetic enzymes and regulatory proteins can be targeted using small molecules. Histone deacetylase inhibitors (HDACis), as a class of epigenetic drugs, are widely used to treat various cancers and other diseases involving abnormal gene expression. Results Specially, HDACis have emerged as a promising strategy to enhance the therapeutic effect of non-neoplastic conditions, including neurological disorders, cardiovascular diseases, renal diseases, autoimmune diseases, inflammatory diseases, infectious diseases and rare diseases, along with their related mechanisms. However, their clinical efficacy has been limited by drug resistance and toxicity. Conclusions To date, most clinical trials of HDAC inhibitors have been related to the treatment of cancer rather than the treatment of non-cancer diseases, for which experimental studies are gradually underway. Discussions regarding non-neoplastic diseases often concentrate on specific disease types. Therefore, this review highlights the development of HDACis and their potential therapeutic applications in non-neoplastic diseases, either as monotherapy or in combination with other drugs or therapies.
Collapse
Affiliation(s)
- Chunxiao Zhou
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Dengke Zhao
- Harbin Medical University, Harbin, 150000, China
| | - Chunyan Wu
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Zhimin Wu
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Wen Zhang
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Shilv Chen
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Xindong Zhao
- College of Medicine, Qingdao University, Qingdao, 266000, China
| | - Shaoling Wu
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| |
Collapse
|
8
|
Verma A, Bhagchandani T, Rai A, Nikita, Sardarni UK, Bhavesh NS, Gulati S, Malik R, Tandon R. Short-Chain Fatty Acid (SCFA) as a Connecting Link between Microbiota and Gut-Lung Axis-A Potential Therapeutic Intervention to Improve Lung Health. ACS OMEGA 2024; 9:14648-14671. [PMID: 38585101 PMCID: PMC10993281 DOI: 10.1021/acsomega.3c05846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 04/09/2024]
Abstract
The microbiome is an integral part of the human gut, and it plays a crucial role in the development of the immune system and homeostasis. Apart from the gut microbiome, the airway microbial community also forms a distinct and crucial part of the human microbiota. Furthermore, several studies indicate the existence of communication between the gut microbiome and their metabolites with the lung airways, called "gut-lung axis". Perturbations in gut microbiota composition, termed dysbiosis, can have acute and chronic effects on the pathophysiology of lung diseases. Microbes and their metabolites in lung stimulate various innate immune pathways, which modulate the expression of the inflammatory genes in pulmonary leukocytes. For instance, gut microbiota-derived metabolites such as short-chain fatty acids can suppress lung inflammation through the activation of G protein-coupled receptors (free fatty acid receptors) and can also inhibit histone deacetylase, which in turn influences the severity of acute and chronic respiratory diseases. Thus, modulation of the gut microbiome composition through probiotic/prebiotic usage and fecal microbiota transplantation can lead to alterations in lung homeostasis and immunity. The resulting manipulation of immune cells function through microbiota and their key metabolites paves the way for the development of novel therapeutic strategies in improving the lung health of individuals affected with various lung diseases including SARS-CoV-2. This review will shed light upon the mechanistic aspect of immune system programming through gut and lung microbiota and exploration of the relationship between gut-lung microbiome and also highlight the therapeutic potential of gut microbiota-derived metabolites in the management of respiratory diseases.
Collapse
Affiliation(s)
- Anjali Verma
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Tannu Bhagchandani
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ankita Rai
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nikita
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Urvinder Kaur Sardarni
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Neel Sarovar Bhavesh
- Transcription
Regulation Group, International Centre for
Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Sameer Gulati
- Department
of Medicine, Lady Hardinge Medical College
(LHMC), New Delhi 110058, India
| | - Rupali Malik
- Department
of Medicine, Vardhman Mahavir Medical College
and Safdarjung Hospital, New Delhi 110029, India
| | - Ravi Tandon
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
9
|
Stakišaitis D, Kapočius L, Tatarūnas V, Gečys D, Mickienė A, Tamošuitis T, Ugenskienė R, Vaitkevičius A, Balnytė I, Lesauskaitė V. Effects of Combined Treatment with Sodium Dichloroacetate and Sodium Valproate on the Genes in Inflammation- and Immune-Related Pathways in T Lymphocytes from Patients with SARS-CoV-2 Infection with Pneumonia: Sex-Related Differences. Pharmaceutics 2024; 16:409. [PMID: 38543303 PMCID: PMC10974540 DOI: 10.3390/pharmaceutics16030409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 12/10/2024] Open
Abstract
The study presents data on the anti-inflammatory effects of a combination of sodium dichloroacetate and sodium valproate (DCA-VPA) on the expression of inflammation- and immune response-related genes in T lymphocytes of SARS-CoV-2 patients. The study aimed to assess the effects of DCA-VPA on the genes of cytokine activity, chemokine-mediated signaling, neutrophil chemotaxis, lymphocyte chemotaxis, T-cell chemotaxis, and regulation of T-cell proliferation pathways. The study included 21 patients with SARS-CoV-2 infection and pneumonia: 9 male patients with a mean age of 68.44 ± 15.32 years and 12 female patients with a mean age of 65.42 ± 15.74 years. They were hospitalized between December 2022 and March 2023. At the time of testing, over 90% of sequences analyzed in Lithuania were found to be of the omicron variant of SARS-CoV-2. The T lymphocytes from patients were treated with 5 mmol DCA and 2 mmol VPA for 24 h in vitro. The effect of the DCA-VPA treatment on gene expression in T lymphocytes was analyzed via gene sequencing. The study shows that DCA-VPA has significant anti-inflammatory effects and apparent sex-related differences. The effect is more potent in T cells from male patients with SARS-CoV-2 infection and pneumonia than in females.
Collapse
Affiliation(s)
- Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (I.B.)
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania
| | - Linas Kapočius
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (I.B.)
| | - Vacis Tatarūnas
- Institute of Cardiology, Laboratory of Molecular Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.T.); (D.G.); (V.L.)
| | - Dovydas Gečys
- Institute of Cardiology, Laboratory of Molecular Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.T.); (D.G.); (V.L.)
| | - Auksė Mickienė
- Department of Infectious Diseases, Lithuanian University of Health Sciences, 47116 Kaunas, Lithuania;
| | - Tomas Tamošuitis
- Department of Intensive Care Medicine, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania;
| | - Rasa Ugenskienė
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania;
| | - Arūnas Vaitkevičius
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University Hospital Santaros Klinikos, Vilnius University, 08661 Vilnius, Lithuania;
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (I.B.)
| | - Vaiva Lesauskaitė
- Institute of Cardiology, Laboratory of Molecular Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.T.); (D.G.); (V.L.)
| |
Collapse
|
10
|
Trionfetti F, Alonzi T, Bontempi G, Terri M, Battistelli C, Montaldo C, Repele F, Rotili D, Valente S, Zwergel C, Matusali G, Maggi F, Goletti D, Tripodi M, Mai A, Strippoli R. HDAC1-3 inhibition increases SARS-CoV-2 replication and productive infection in lung mesothelial and epithelial cells. Front Cell Infect Microbiol 2023; 13:1257683. [PMID: 38162580 PMCID: PMC10757821 DOI: 10.3389/fcimb.2023.1257683] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/30/2023] [Indexed: 01/03/2024] Open
Abstract
Background Despite the significant progress achieved in understanding the pathology and clinical management of SARS-CoV-2 infection, still pathogenic and clinical issues need to be clarified. Treatment with modulators of epigenetic targets, i.e., epidrugs, is a current therapeutic option in several cancers and could represent an approach in the therapy of viral diseases. Results Aim of this study was the analysis of the role of histone deacetylase (HDAC) inhibition in the modulation of SARS-CoV-2 infection of mesothelial cells (MCs).MeT5A cells, a pleura MC line, were pre-treated with different specific class I and IIb HDAC inhibitors. Unexpectedly, treatment with HDAC1-3 inhibitors significantly increased ACE2/TMPRSS2 expression, suggesting a role in favoring SARS-CoV-2 infection. We focused our analysis on the most potent ACE2/TMPRSS2 inducer among the inhibitors analysed, MS-275, a HDAC1-3 inhibitor. ACE2/TMPRSS2 expression was validated by Western Blot (WB) and immunofluorescence. The involvement of HDAC inhibition in receptor induction was confirmed by HDAC1/HDAC2 silencing. In accordance to the ACE2/TMPRSS2 expression data, MS-275 increased SARS-CoV-2 replication and virus propagation in Vero E6 cells.Notably, MS-275 was able to increase ACE2/TMPRSS2 expression and SARS-CoV-2 production, although to a lesser extent, also in the lung adenocarcinoma cell line Calu-3 cells.Mechanistically, treatment with MS-275 increased H3 and H4 histone acetylation at ACE2/TMPRSS2 promoters, increasing their transcription. Conclusion This study highlights a previously unrecognized effect of HDAC1-3 inhibition in increasing SARS-CoV-2 cell entry, replication and productive infection correlating with increased expression of ACE2 and TMPRSS2. These data, while adding basic insight into COVID-19 pathogenesis, warn for the use of HDAC inhibitors in SARS-CoV-2 patients.
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
| | - Tonino Alonzi
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-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
| | | | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Federica Repele
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Giulia Matusali
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, Rome, Italy
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, 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
|
11
|
Zou S, Li X, Huang Y, Zhang B, Tang H, Xue Y, Zheng Y. Properties and biotechnological applications of microbial deacetylase. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12613-1. [PMID: 37326683 DOI: 10.1007/s00253-023-12613-1] [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: 03/10/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023]
Abstract
Deacetylases, a class of enzymes that can catalyze the hydrolysis of acetylated substrates to remove the acetyl group, used in producing various products with high qualities, are one of the most influential industrial enzymes. These enzymes are highly specific, non-toxic, sustainable, and eco-friendly biocatalysts. Deacetylases and deacetylated compounds have been widely applicated in pharmaceuticals, medicine, food, and the environment. This review synthetically summarizes deacetylases' sources, characterizations, classifications, and applications. Moreover, the typical structural characteristics of deacetylases from different microbial sources are summarized. We also reviewed the deacetylase-catalyzed reactions for producing various deacetylated compounds, such as chitosan-oligosaccharide (COS), mycothiol, 7-aminocephalosporanic acid (7-ACA), glucosamines, amino acids, and polyamines. It is aimed to expound on the advantages and challenges of deacetylases in industrial applications. Moreover, it also serves perspectives on obtaining promising and innovative biocatalysts for enzymatic deacetylation. KEYPOINTS: • The fundamental properties of microbial deacetylases of various microorganisms are presented. • The biochemical characterizations, structures, and catalyzation mechanisms of microbial deacetylases are summarized. • The applications of microbial deacetylases in food, pharmaceutical, medicine, and the environment were discussed.
Collapse
Affiliation(s)
- Shuping Zou
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Xia Li
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Yinfeng Huang
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Bing Zhang
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Heng Tang
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Yaping Xue
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China.
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China.
| | - Yuguo Zheng
- National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| |
Collapse
|
12
|
Luo G, Liu B, Fu T, Liu Y, Li B, Li N, Geng Q. The Role of Histone Deacetylases in Acute Lung Injury-Friend or Foe. Int J Mol Sci 2023; 24:ijms24097876. [PMID: 37175583 PMCID: PMC10178380 DOI: 10.3390/ijms24097876] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023] Open
Abstract
Acute lung injury (ALI), caused by intrapulmonary or extrapulmonary factors such as pneumonia, shock, and sepsis, eventually disrupts the alveolar-capillary barrier, resulting in diffuse pulmonary oedema and microatasis, manifested by refractory hypoxemia, and respiratory distress. Not only is ALI highly lethal, but even if a patient survives, there are also multiple sequelae. Currently, there is no better treatment than supportive care, and we urgently need to find new targets to improve ALI. Histone deacetylases (HDACs) are epigenetically important enzymes that, together with histone acetylases (HATs), regulate the acetylation levels of histones and non-histones. While HDAC inhibitors (HDACis) play a therapeutic role in cancer, inflammatory, and neurodegenerative diseases, there is also a large body of evidence suggesting the potential of HDACs as therapeutic targets in ALI. This review explores the unique mechanisms of HDACs in different cell types of ALI, including macrophages, pulmonary vascular endothelial cells (VECs), alveolar epithelial cells (AECs), and neutrophils.
Collapse
Affiliation(s)
- Guoqing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tinglv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Boyang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
13
|
Fabbrizi E, Fiorentino F, Carafa V, Altucci L, Mai A, Rotili D. Emerging Roles of SIRT5 in Metabolism, Cancer, and SARS-CoV-2 Infection. Cells 2023; 12:cells12060852. [PMID: 36980194 PMCID: PMC10047932 DOI: 10.3390/cells12060852] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Sirtuin 5 (SIRT5) is a predominantly mitochondrial enzyme catalyzing the removal of glutaryl, succinyl, malonyl, and acetyl groups from lysine residues through a NAD+-dependent deacylase mechanism. SIRT5 is an important regulator of cellular homeostasis and modulates the activity of proteins involved in different metabolic pathways such as glycolysis, tricarboxylic acid (TCA) cycle, fatty acid oxidation, electron transport chain, generation of ketone bodies, nitrogenous waste management, and reactive oxygen species (ROS) detoxification. SIRT5 controls a wide range of aspects of myocardial energy metabolism and plays critical roles in heart physiology and stress responses. Moreover, SIRT5 has a protective function in the context of neurodegenerative diseases, while it acts as a context-dependent tumor promoter or suppressor. In addition, current research has demonstrated that SIRT5 is implicated in the SARS-CoV-2 infection, although opposing conclusions have been drawn in different studies. Here, we review the current knowledge on SIRT5 molecular actions under both healthy and diseased settings, as well as its functional effects on metabolic targets. Finally, we revise the potential of SIRT5 as a therapeutic target and provide an overview of the currently reported SIRT5 modulators, which include both activators and inhibitors.
Collapse
Affiliation(s)
- Emanuele Fabbrizi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Vincenzo Carafa
- Department of Precision Medicine, Università degli Studi della Campania “L. Vanvitelli”, 80138 Naples, Italy
- BIOGEM, 83031 Ariano Irpino, Italy
| | - Lucia Altucci
- Department of Precision Medicine, Università degli Studi della Campania “L. Vanvitelli”, 80138 Naples, Italy
- BIOGEM, 83031 Ariano Irpino, Italy
- IEOS—Istituto per l’Endocrinologia e Oncologia Sperimentale, CNR, 80131 Naples, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
- Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (A.M.); (D.R.); Tel.: +39-0649913392 (A.M.); +39-0649913237 (D.R.); Fax: +39-0649693268 (A.M.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (A.M.); (D.R.); Tel.: +39-0649913392 (A.M.); +39-0649913237 (D.R.); Fax: +39-0649693268 (A.M.)
| |
Collapse
|
14
|
Zahid S, Ali Y, Rashid S. Structural-based design of HD-TAC7 PROteolysis TArgeting chimeras (PROTACs) candidate transformations to abrogate SARS-CoV-2 infection. J Biomol Struct Dyn 2023; 41:14566-14581. [PMID: 36841549 DOI: 10.1080/07391102.2023.2183037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/16/2023] [Indexed: 02/27/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for about 672 million infections and 6.85 million deaths worldwide. Upon SARS-CoV-2 infection, Histone deacetylases (HDACs) hyperactivate the pro-inflammatory response resulting in stimulation of Acetyl-Coenzyme A and cholesterol for viral entry. HDAC3 inhibition results in the anti-inflammatory activity and reduction of pro-inflammatory cytokines that may restrict COVID-19 progression. Here, we have designed 44 conformational ensembles of previously known HD-TAC7 by enumerating torsions of dihedral angles tested for their binding preferences against HDAC3. Through scrutinizing their placements at active site and binding affinities, three hits were isolated. Cereblon (CRBN) is a well-known E3 ligase that facilitates Proteolysis Targeting Chimeras (PROTACs) targeting. Three entities, including HDAC3-binding moiety (4-acetamido-N-(2-amino-4 fluorophenyl) benzamide), a 6-carbon linker, and CRBN binding ligand (pomalidomide) were assembled to design 4 PROTACs followed by energy minimization and docking against HDAC3 and CRBN, respectively. Subsequent molecular dynamics (MD) and free energy analyses corroborated similar binding trends and favorable energy values. Among all cases, Met88, GLu106, Pro352, Trp380 and Trp388 residues of CRBN, and Pro23, Arg28, Lys194, Phe199, Leu266, Thr299 and Ile346 residues of HDAC3 were engaged in PROTAC binding. Thus, conformational dynamics of both HDAC3 and CRBN moieties are essential for the placement of PROTAC, resulting in target degradation. Overall, the proposed bifunctional small molecules may effectively target HDAC3, stimulating innate immune response to restrict COVID-19 hyperinflammation. This study supports the basis for designing new PROTACs by limiting the conformational search space that may prove more efficient for targeting the protein of interest.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sana Zahid
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Yasir Ali
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sajid Rashid
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
15
|
Kiss-Szemán AJ, Takács L, Orgován Z, Stráner P, Jákli I, Schlosser G, Masiulis S, Harmat V, Menyhárd DK, Perczel A. A carbapenem antibiotic inhibiting a mammalian serine protease: structure of the acylaminoacyl peptidase-meropenem complex. Chem Sci 2022; 13:14264-14276. [PMID: 36545146 PMCID: PMC9749117 DOI: 10.1039/d2sc05520a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/06/2022] [Indexed: 11/10/2022] Open
Abstract
The structure of porcine AAP (pAAP) in a covalently bound complex with meropenem was determined by cryo-EM to 2.1 Å resolution, showing the mammalian serine-protease inhibited by a carbapenem antibiotic. AAP is a modulator of the ubiquitin-proteasome degradation system and the site of a drug-drug interaction between the widely used antipsychotic, valproate and carbapenems. The active form of pAAP - a toroidal tetramer - binds four meropenem molecules covalently linked to the catalytic Ser587 of the serine-protease triad, in an acyl-enzyme state. AAP is hindered from fully processing the antibiotic by the displacement and protonation of His707 of the catalytic triad. We show that AAP is made susceptible to the association by its unusually sheltered active pockets and flexible catalytic triads, while the carbapenems possess sufficiently small substituents on their β-lactam rings to fit into the shallow substrate-specificity pocket of the enzyme.
Collapse
Affiliation(s)
- Anna J Kiss-Szemán
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University Pázmány Péter sétány 1/A Budapest Hungary
| | - Luca Takács
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University Pázmány Péter sétány 1/A Budapest Hungary
| | - Zoltán Orgován
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences Budapest Hungary
| | - Pál Stráner
- ELKH-ELTE Protein Modelling Research Group, Eötvös Loránd Research Network Budapest Hungary +36-1-372-2500/1653 +36-1-372-2500/6547
| | - Imre Jákli
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University Pázmány Péter sétány 1/A Budapest Hungary
- ELKH-ELTE Protein Modelling Research Group, Eötvös Loránd Research Network Budapest Hungary +36-1-372-2500/1653 +36-1-372-2500/6547
| | - Gitta Schlosser
- ELKH-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Institute of Chemistry, Eötvös Loránd University Budapest Hungary
| | - Simonas Masiulis
- Materials and Structural Analysis Division, Thermo Fisher Scientific Eindhoven The Netherlands
| | - Veronika Harmat
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University Pázmány Péter sétány 1/A Budapest Hungary
- ELKH-ELTE Protein Modelling Research Group, Eötvös Loránd Research Network Budapest Hungary +36-1-372-2500/1653 +36-1-372-2500/6547
| | - Dóra K Menyhárd
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University Pázmány Péter sétány 1/A Budapest Hungary
- ELKH-ELTE Protein Modelling Research Group, Eötvös Loránd Research Network Budapest Hungary +36-1-372-2500/1653 +36-1-372-2500/6547
| | - András Perczel
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University Pázmány Péter sétány 1/A Budapest Hungary
- ELKH-ELTE Protein Modelling Research Group, Eötvös Loránd Research Network Budapest Hungary +36-1-372-2500/1653 +36-1-372-2500/6547
| |
Collapse
|
16
|
Zhang S, Meng Y, Zhou L, Qiu L, Wang H, Su D, Zhang B, Chan K, Han J. Targeting epigenetic regulators for inflammation: Mechanisms and intervention therapy. MedComm (Beijing) 2022; 3:e173. [PMID: 36176733 PMCID: PMC9477794 DOI: 10.1002/mco2.173] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/28/2022] [Accepted: 08/05/2022] [Indexed: 11/11/2022] Open
Abstract
Emerging evidence indicates that resolution of inflammation is a critical and dynamic endogenous process for host tissues defending against external invasive pathogens or internal tissue injury. It has long been known that autoimmune diseases and chronic inflammatory disorders are characterized by dysregulated immune responses, leading to excessive and uncontrol tissue inflammation. The dysregulation of epigenetic alterations including DNA methylation, posttranslational modifications to histone proteins, and noncoding RNA expression has been implicated in a host of inflammatory disorders and the immune system. The inflammatory response is considered as a critical trigger of epigenetic alterations that in turn intercede inflammatory actions. Thus, understanding the molecular mechanism that dictates the outcome of targeting epigenetic regulators for inflammatory disease is required for inflammation resolution. In this article, we elucidate the critical role of the nuclear factor-κB signaling pathway, JAK/STAT signaling pathway, and the NLRP3 inflammasome in chronic inflammatory diseases. And we formulate the relationship between inflammation, coronavirus disease 2019, and human cancers. Additionally, we review the mechanism of epigenetic modifications involved in inflammation and innate immune cells. All that matters is that we propose and discuss the rejuvenation potential of interventions that target epigenetic regulators and regulatory mechanisms for chronic inflammation-associated diseases to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Su Zhang
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Yang Meng
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Lian Zhou
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Lei Qiu
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Heping Wang
- Department of NeurosurgeryTongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dan Su
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Bo Zhang
- Laboratory of Cancer Epigenetics and GenomicsDepartment of Gastrointestinal SurgeryFrontiers Science Center for Disease‐Related Molecular NetworkWest China HospitalSichuan UniversityChengduChina
| | - Kui‐Ming Chan
- Department of Biomedical SciencesCity University of Hong KongHong KongChina
| | - Junhong Han
- Laboratory of Cancer Epigenetics and GenomicsFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
17
|
Efficacy of Valproic Acid Against Coronavirus Disease 2019 Infection or Severity: A Pilot Study. Clin Neuropharmacol 2022; 45:175-176. [DOI: 10.1097/wnf.0000000000000529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
18
|
Lu Y, Zhao N, Du Y. Comprehensive bioinformatics analysis reveals common potential mechanisms, progression markers, and immune cells of coronary virus disease 2019 and atrial fibrillation. Front Cardiovasc Med 2022; 9:1027026. [PMID: 36352845 PMCID: PMC9637541 DOI: 10.3389/fcvm.2022.1027026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/07/2022] [Indexed: 12/01/2023] Open
Abstract
OBJECTIVES Atrial fibrillation (AF) is the most common arrhythmia in coronary virus disease 2019 (COVID-19) patients, especially in severe patients. A history of AF can exacerbate COVID-19 symptoms. COVID-19 Patients with new-onset AF have prolonged hospital stays and increased death risk. However, the mechanisms and targets of the interaction between COVID-19 and AF have not been elucidated. MATERIALS AND METHODS We used a series of bioinformatics analyses to understand biological pathways, protein-protein interaction (PPI) networks, gene regulatory networks (GRNs), and protein-chemical interactions between COVID-19 and AF and constructed an AF-related gene signature to assess COVID-19 severity and prognosis. RESULTS We found folate and one-carbon metabolism, calcium regulation, and TFG-β signaling pathway as potential mechanisms linking COVID-19 and AF, which may be involved in alterations in neutrophil metabolism, inflammation, and endothelial cell function. We identified hug genes and found that NF-κb, hsa-miR-1-3p, hsa-miR-124-3p, valproic acid, and quercetin may be key regulatory molecules. We constructed a 3-gene signature consisting of ARG1, GIMAP7, and RFX2 models for the assessment of COVID-19 severity and prognosis, and found that they are associated with neutrophils, T cells, and hematopoietic stem cells, respectively. CONCLUSION Our study reveals a dysregulation of metabolism, inflammation, and immunity between COVID-19 and AF, and identified several therapeutic targets and progression markers. We hope that the results will reveal important insights into the complex interactions between COVID-19 and AF that will drive novel drug development and help in severity assessment.
Collapse
Affiliation(s)
- Yang Lu
- Department of Cardiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Research Center of Ion Channelopathy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Tongji Medical College, Union Hospital, Institute of Cardiology, Huazhong University of Science and Technology, Wuhan, China
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ning Zhao
- Department of Cardiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Research Center of Ion Channelopathy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Tongji Medical College, Union Hospital, Institute of Cardiology, Huazhong University of Science and Technology, Wuhan, China
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yimei Du
- Department of Cardiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Research Center of Ion Channelopathy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Tongji Medical College, Union Hospital, Institute of Cardiology, Huazhong University of Science and Technology, Wuhan, China
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Long-term analysis of the effects of COVID-19 in people with epilepsy: Results from a multicenter on-line survey across the pandemic waves. Epilepsy Behav 2022; 135:108900. [PMID: 36115083 PMCID: PMC9404228 DOI: 10.1016/j.yebeh.2022.108900] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/12/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022]
Abstract
PURPOSE The worldwide pandemic caused by SARS-CoV-2 virus posed many challenges to the scientific and medical communities, including the protection and management of fragile populations. People with epilepsy (PWE) are a heterogenous group of subjects, with different treatment regimens and severity of symptoms. During the National lockdown, in Italy many patients with chronic conditions lost their regular follow-up program. The aim of this study was to investigate the impact of COVID-19 on their health status, from the start of the pandemic (March 2020) to July 2021 and one year later. METHODS We proposed an online questionnaire to subjects followed up at different epilepsy centers located in Milano, Monza & Lodi, three of Lombardy, Northern Italy, the most affected areas by the pandemic. Survey evaluated age, sex, characteristics of patients, type of epilepsy and therapies, COVID-19 diagnosis, vaccines, sleep quality, and anxiety status. RESULTS Among 178 analyzed surveys, 37 individuals reported symptoms of COVID-19 in closed contacts, including 9 with molecular diagnosis and 16 PWE performing the nasopharyngeal swab with 3 positive cases. One year later, 35 individuals reported at least one symptom overlapping with those typical of COVID-19, 8 received COVID-19 diagnosis, among which 6 were positive for SARS-CoV-2 infection. According to the sleep quality scale assessment, most PWE (52.3%) had poor sleep quality. Assessing anxiety status, 32 (38.1%) had a pathological score. CONCLUSION In this multicenter study, we observed that PWE do not appear to be at a higher risk of severe COVID-19. It will be fundamental monitoring this group to assess possible differences in long-COVID-19 and/or neuro-COVID-19 prevalence. On the other hand, our survey confirmed the impact of the pandemic on anxiety and quality of sleep in PWE. Thus, it is important to promptly recognize and treat psychological distress in PWE, because it could be a risk factor in seizure aggravation and quality-of-life deterioration. Telemedicine appears to be a useful tool to support patients with chronic diseases, such as epilepsy.
Collapse
|
20
|
Wang T, Cao Y, Zhang H, Wang Z, Man CH, Yang Y, Chen L, Xu S, Yan X, Zheng Q, Wang Y. COVID-19 metabolism: Mechanisms and therapeutic targets. MedComm (Beijing) 2022; 3:e157. [PMID: 35958432 PMCID: PMC9363584 DOI: 10.1002/mco2.157] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 01/18/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) dysregulates antiviral signaling, immune response, and cell metabolism in human body. Viral genome and proteins hijack host metabolic network to support viral biogenesis and propagation. However, the regulatory mechanism of SARS-CoV-2-induced metabolic dysfunction has not been elucidated until recently. Multiomic studies of coronavirus disease 2019 (COVID-19) revealed an intensive interaction between host metabolic regulators and viral proteins. SARS-CoV-2 deregulated cellular metabolism in blood, intestine, liver, pancreas, fat, and immune cells. Host metabolism supported almost every stage of viral lifecycle. Strikingly, viral proteins were found to interact with metabolic enzymes in different cellular compartments. Biochemical and genetic assays also identified key regulatory nodes and metabolic dependencies of viral replication. Of note, cholesterol metabolism, lipid metabolism, and glucose metabolism are broadly involved in viral lifecycle. Here, we summarized the current understanding of the hallmarks of COVID-19 metabolism. SARS-CoV-2 infection remodels host cell metabolism, which in turn modulates viral biogenesis and replication. Remodeling of host metabolism creates metabolic vulnerability of SARS-CoV-2 replication, which could be explored to uncover new therapeutic targets. The efficacy of metabolic inhibitors against COVID-19 is under investigation in several clinical trials. Ultimately, the knowledge of SARS-CoV-2-induced metabolic reprogramming would accelerate drug repurposing or screening to combat the COVID-19 pandemic.
Collapse
Affiliation(s)
- Tianshi Wang
- Shanghai Key Laboratory for Tumor Microenvironment and InflammationDepartment of Biochemistry and Molecular Cell BiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Cao
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Haiyan Zhang
- Bai Jia Obstetrics and Gynecology HospitalShanghaiChina
| | - Zihao Wang
- Fudan University Shanghai Cancer CenterKey Laboratory of Breast Cancer in ShanghaiShanghai Key Laboratory of Radiation OncologyCancer Instituteand The Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- The International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghaiChina
| | - Cheuk Him Man
- Division of HematologyDepartment of MedicineUniversity of Hong KongPokfulamHong Kong, China
| | - Yunfan Yang
- Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Lingchao Chen
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersShanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationNeurosurgical Institute of Fudan UniversityShanghai Clinical Medical Center of NeurosurgeryShanghaiChina
| | - Shuangnian Xu
- Department of HematologySouthwest HospitalArmy Medical UniversityChongqingChina
| | - Xiaojing Yan
- Department of HematologyThe First Affiliated Hospital of China Medical UniversityShenyangChina
| | - Quan Zheng
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Ping Wang
- Fudan University Shanghai Cancer CenterKey Laboratory of Breast Cancer in ShanghaiShanghai Key Laboratory of Radiation OncologyCancer Instituteand The Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- The International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghaiChina
| |
Collapse
|
21
|
Li Z, Fang P, Duan P, Chen J, Fang L, Xiao S. Porcine Deltacoronavirus Infection Cleaves HDAC2 to Attenuate Its Antiviral Activity. J Virol 2022; 96:e0102722. [PMID: 35916536 PMCID: PMC9400482 DOI: 10.1128/jvi.01027-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 02/04/2023] Open
Abstract
Protein acetylation plays an important role during virus infection. Thus, it is not surprising that viruses always evolve elaborate mechanisms to regulate the functions of histone deacetylases (HDACs), the essential transcriptional and epigenetic regulators for deacetylation. Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, causes severe diarrhea in suckling piglets and has the potential to infect humans. In this study, we found that PDCoV infection inhibited cellular HDAC activity. By screening the expressions of different HDAC subfamilies after PDCoV infection, we unexpectedly found that HDAC2 was cleaved. Ectopic expression of HDAC2 significantly inhibited PDCoV replication, while the reverse effects could be observed after treatment with an HDAC2 inhibitor (CAY10683) or the knockdown of HDAC2 expression by specific siRNA. Furthermore, we demonstrated that PDCoV-encoded nonstructural protein 5 (nsp5), a 3C-like protease, was responsible for HDAC2 cleavage through its protease activity. Detailed analyses showed that PDCoV nsp5 cleaved HDAC2 at glutamine 261 (Q261), and the cleaved fragments (amino acids 1 to 261 and 262 to 488) lost the ability to inhibit PDCoV replication. Interestingly, the Q261 cleavage site is highly conserved in HDAC2 homologs from other mammalian species, and the nsp5s encoded by seven tested mammalian coronaviruses also cleaved HDAC2, suggesting that cleaving HDAC2 may be a common strategy used by different mammalian coronaviruses to antagonize the antiviral role of HDAC2. IMPORTANCE As an emerging porcine enteropathogenic coronavirus that possesses the potential to infect humans, porcine deltacoronavirus (PDCoV) is receiving increasing attention. In this work, we found that PDCoV infection downregulated cellular histone deacetylase (HDAC) activity. Of particular interest, the viral 3C-like protease, encoded by the PDCoV nonstructural protein 5 (nsp5), cleaved HDAC2, and this cleavage could be observed in the context of PDCoV infection. Furthermore, the cleavage of HDAC2 appears to be a common strategy among mammalian coronaviruses, including the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), to antagonize the antiviral role of HDAC2. To our knowledge, PDCoV nsp5 is the first identified viral protein that can cleave cellular HDAC2. Results from our study provide new targets to develop drugs combating coronavirus infection.
Collapse
Affiliation(s)
- Zhuang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Puxian Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Panpan Duan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Jiyao Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Liurong Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Shaobo Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
22
|
The global succinylation of SARS-CoV-2–infected host cells reveals drug targets. Proc Natl Acad Sci U S A 2022; 119:e2123065119. [PMID: 35858407 PMCID: PMC9335334 DOI: 10.1073/pnas.2123065119] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The continued rapid evolution of SARS-CoV-2 and the emergence of immune-evading variants pose significant challenges to COVID-19 prevention and control, highlighting the urgent need for development of novel antiviral therapies. Our study found that SARS-CoV-2 infection promotes host succinylation and inhibits several key enzymes of the TCA, a crucial metabolic pathway that connects carbohydrate, fat, and protein metabolism, as well as regulating cellular energy. Additionally, viral NSP14 is capable to participate in succinylation through interacting with host SIRT5, a cellular desuccinylase. It is noteworthy that succinylation inhibitors can significantly reduce the viral replication, as a potential guide for the treatment of COVID-19. SARS-CoV-2, the causative agent of the COVID-19 pandemic, undergoes continuous evolution, highlighting an urgent need for development of novel antiviral therapies. Here we show a quantitative mass spectrometry-based succinylproteomics analysis of SARS-CoV-2 infection in Caco-2 cells, revealing dramatic reshape of succinylation on host and viral proteins. SARS-CoV-2 infection promotes succinylation of several key enzymes in the TCA, leading to inhibition of cellular metabolic pathways. We demonstrated that host protein succinylation is regulated by viral nonstructural protein (NSP14) through interaction with sirtuin 5 (SIRT5); overexpressed SIRT5 can effectively inhibit virus replication. We found succinylation inhibitors possess significant antiviral effects. We also found that SARS-CoV-2 nucleocapsid and membrane proteins underwent succinylation modification, which was conserved in SARS-CoV-2 and its variants. Collectively, our results uncover a regulatory mechanism of host protein posttranslational modification and cellular pathways mediated by SARS-CoV-2, which may become antiviral drug targets against COVID-19.
Collapse
|
23
|
Ripamonti C, Spadotto V, Pozzi P, Stevenazzi A, Vergani B, Marchini M, Sandrone G, Bonetti E, Mazzarella L, Minucci S, Steinkühler C, Fossati G. HDAC Inhibition as Potential Therapeutic Strategy to Restore the Deregulated Immune Response in Severe COVID-19. Front Immunol 2022; 13:841716. [PMID: 35592335 PMCID: PMC9111747 DOI: 10.3389/fimmu.2022.841716] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/01/2022] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has had a devastating impact worldwide and has been a great challenge for the scientific community. Vaccines against SARS-CoV-2 are now efficiently lessening COVID-19 mortality, although finding a cure for this infection is still a priority. An unbalanced immune response and the uncontrolled release of proinflammatory cytokines are features of COVID-19 pathophysiology and contribute to disease progression and worsening. Histone deacetylases (HDACs) have gained interest in immunology, as they regulate the innate and adaptative immune response at different levels. Inhibitors of these enzymes have already proven therapeutic potential in cancer and are currently being investigated for the treatment of autoimmune diseases. We thus tested the effects of different HDAC inhibitors, with a focus on a selective HDAC6 inhibitor, on immune and epithelial cells in in vitro models that mimic cells activation after viral infection. Our data indicate that HDAC inhibitors reduce cytokines release by airway epithelial cells, monocytes and macrophages. This anti-inflammatory effect occurs together with the reduction of monocytes activation and T cell exhaustion and with an increase of T cell differentiation towards a T central memory phenotype. Moreover, HDAC inhibitors hinder IFN-I expression and downstream effects in both airway epithelial cells and immune cells, thus potentially counteracting the negative effects promoted in critical COVID-19 patients by the late or persistent IFN-I pathway activation. All these data suggest that an epigenetic therapeutic approach based on HDAC inhibitors represents a promising pharmacological treatment for severe COVID-19 patients.
Collapse
Affiliation(s)
- Chiara Ripamonti
- New Drug Incubator Department, Italfarmaco Group, Cinisello Balsamo, Italy
| | - Valeria Spadotto
- New Drug Incubator Department, Italfarmaco Group, Cinisello Balsamo, Italy
| | - Pietro Pozzi
- New Drug Incubator Department, Italfarmaco Group, Cinisello Balsamo, Italy
| | - Andrea Stevenazzi
- New Drug Incubator Department, Italfarmaco Group, Cinisello Balsamo, Italy
| | - Barbara Vergani
- New Drug Incubator Department, Italfarmaco Group, Cinisello Balsamo, Italy
| | - Mattia Marchini
- New Drug Incubator Department, Italfarmaco Group, Cinisello Balsamo, Italy
| | - Giovanni Sandrone
- New Drug Incubator Department, Italfarmaco Group, Cinisello Balsamo, Italy
| | - Emanuele Bonetti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Luca Mazzarella
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | | | - Gianluca Fossati
- New Drug Incubator Department, Italfarmaco Group, Cinisello Balsamo, Italy
| |
Collapse
|
24
|
Nemani K, Williams SZ, Olfson M, Leckman-Westin E, Finnerty M, Kammer J, Smith TE, Silverman DJ, Lindenmayer JP, Capichioni G, Clelland J, Goff DC. Association Between the Use of Psychotropic Medications and the Risk of COVID-19 Infection Among Long-term Inpatients With Serious Mental Illness in a New York State-wide Psychiatric Hospital System. JAMA Netw Open 2022; 5:e2210743. [PMID: 35522282 PMCID: PMC9077485 DOI: 10.1001/jamanetworkopen.2022.10743] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
IMPORTANCE Individuals with serious mental illness are at increased risk of severe COVID-19 infection. Several psychotropic medications have been identified as potential therapeutic agents to prevent or treat COVID-19 but have not been systematically examined in this population. OBJECTIVE To evaluate the associations between the use of psychotropic medications and the risk of COVID-19 infection among adults with serious mental illness receiving long-term inpatient psychiatric treatment. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort study assessed adults with serious mental illness hospitalized in a statewide psychiatric hospital system in New York between March 8 and July 1, 2020. The final date of follow-up was December 1, 2020. The study included 1958 consecutive adult inpatients with serious mental illness (affective or nonaffective psychoses) who received testing for SARS-CoV-2 by reverse transcriptase-polymerase chain reaction or antinucleocapsid antibodies and were continuously hospitalized from March 8 until medical discharge or July 1, 2020. EXPOSURES Psychotropic medications prescribed prior to COVID-19 testing. MAIN OUTCOMES AND MEASURES COVID-19 infection was the primary outcome, defined by a positive SARS-CoV-2 reverse transcriptase-polymerase chain reaction or antibody test result. The secondary outcome was COVID-19-related death among patients with laboratory-confirmed infection. RESULTS Of the 2087 adult inpatients with serious mental illness continuously hospitalized during the study period, 1958 (93.8%) underwent testing and were included in the study; 1442 (73.6%) were men, and the mean (SD) age was 51.4 (14.3) years. A total of 969 patients (49.5%) had laboratory-confirmed COVID-19 infection that occurred while they were hospitalized; of those, 38 (3.9%) died. The use of second-generation antipsychotic medications, as a class, was associated with decreased odds of infection (odds ratio [OR], 0.62; 95% CI, 0.45-0.86), whereas the use of mood stabilizers was associated with increased odds of infection (OR, 1.23; 95% CI, 1.03-1.47). In a multivariable model of individual medications, the use of paliperidone was associated with decreased odds of infection (OR, 0.59; 95% CI, 0.41-0.84), and the use of valproic acid was associated with increased odds of infection (OR, 1.39; 95% CI, 1.10-1.76). Clozapine use was associated with reduced odds of mortality in unadjusted analyses (unadjusted OR, 0.25; 95% CI, 0.10-0.62; fully adjusted OR, 0.43; 95% CI, 0.17-1.12). CONCLUSIONS AND RELEVANCE In this cohort study of adults hospitalized with serious mental illness, the use of second-generation antipsychotic medications was associated with decreased risk of COVID-19 infection, whereas the use of valproic acid was associated with increased risk. Further research is needed to assess the mechanisms that underlie these findings.
Collapse
Affiliation(s)
- Katlyn Nemani
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
- Department of Psychiatry, New York University Langone Medical Center, New York
| | | | - Mark Olfson
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | | | | | | | | | | | - Jean-Pierre Lindenmayer
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
- Department of Psychiatry, New York University Langone Medical Center, New York
| | - Gillian Capichioni
- Department of Psychiatry, New York University Langone Medical Center, New York
| | - James Clelland
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Donald C. Goff
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
- Department of Psychiatry, New York University Langone Medical Center, New York
| |
Collapse
|
25
|
Effects of naringin and valproate interaction on liver steatosis and dyslipidaemia parameters in male C57BL6 mice. Arh Hig Rada Toksikol 2022; 73:71-82. [PMID: 35390239 PMCID: PMC8999592 DOI: 10.2478/aiht-2022-73-3608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/01/2022] [Indexed: 11/21/2022] Open
Abstract
Valproate is a common antiepileptic drug whose adverse effects include liver steatosis and dyslipidaemia. The aim of our study was to see how natural flavonoid antioxidant naringin would interact with valproate and attenuate these adverse effects. For this reason we treated male C57BL6 mice with a combination of 150 mg/kg of valproate and 25 mg/kg naringin every day for 10 days and compared their serum triglycerides, cholesterol, LDL, HDL, VLDL, and liver PPAR-alpha, PGC-1 alpha, ACOX1, Nrf2, SOD, CAT, GSH, and histological signs of steatosis. Valproate increased lipid peroxidation parameters and caused pronounced microvesicular steatosis throughout the hepatic lobule in all acinar zones, but naringin co-administration limited steatosis to the lobule periphery. In addition, it nearly restored total serum cholesterol, LDL, and triglycerides and liver ACOX1 and MDA to control levels. and upregulated PPAR-alpha and PGC-1 alpha, otherwise severely downregulated by valproate. It also increased SOD activity. All these findings suggest that naringin modulates key lipid metabolism regulators and should further be investigated in this model, either alone or combined with other lipid regulating drugs or molecules.
Collapse
|
26
|
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
|
27
|
Moreno-Pérez O, Merino E, Ramos JM, Rodríguez JC, Diaz C, Mas P, Reus S, Sánchez-Martínez R, Boix V, Chico-Sánchez P, Sánchez-Payá J, Portilla J. [Valproic Acid Could Help in the Fight Against COVID-19: a case-control study]. Neurologia 2022:S0213-4853(22)00014-7. [PMID: 35185237 PMCID: PMC8841207 DOI: 10.1016/j.nrl.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/22/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE There is early evidence about Valproic acid (VPA) antiviral effect. Our aim was to investigate the incidence and severity of SARS-CoV-2 infection in VPA users as compared with the general population. MATERIAL AND METHODS A case-control study nested within a cohort, carried out between March 1 and December 17, 2020. Retrospectively, we identified confirmed SARS-CoV-2 infection patients exposed to VPA in our health department (defined as case). We ascertained VPA regimen (all the time (AT)(292 days) or at least 20% of the study period (notAT)(≥58 days) and if VPA levels were in therapeutic range (ATR) (50-100 mcg/mL) in the last 24 months. We calculated the cumulative incidence of SARS-CoV-2 infection and hospital admission in the cases, comparing it with the general unexposed VPA population (controls). RESULTS During the study period, 6183 PCR+ were detected among 281035 inhabitants, of these, 746 were hospitalized. 691 patients were on VPA notAT and 628 (90.1%) AT. The indication for VPA use was epilepsy in 54.9%. The incidence of PCR+ was 1.736 % (OR 0.785 (95%CI 0.443-1.390) and 1.910 % (OR 0.865 (95%CI 0.488-1.533), on VPA notAT and VPA AT patients, respectively vs. 2.201% in people without VPA regimen. Those patients with VPA ATR had a lower risk of PCR + (OR 0.233 (95%CI 0.057-0.951) notAT; OR 0.218 (95%CI 0.053-0.890) AT). Hospital admission incidence was lower in patient on VPA (OR was 0.543 (95% CI 0.076 to 3.871). CONCLUSION Patients with VPA within the therapeutic range had a reduction of SARS-Cov-2 infection incidence greater than 75%. There is a downward trend in the risk of COVID-19 admission by SARS-CoV-2 in patients on VPA therapy. These findings warrant further investigation.
Collapse
Affiliation(s)
- Oscar Moreno-Pérez
- Endocrinology and Nutrition department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinical Medicine department, Miguel Hernández University, Elche, Spain
| | - Esperanza Merino
- Unit of Infectious Diseases, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Jose Manuel Ramos
- Internal Medicine department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinical Medicine department, Miguel Hernández University, Elche, Spain
| | - Juan Carlos Rodríguez
- Microbiology department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
- Miguel Hernández University, Elche, Spain
| | - Carmina Diaz
- Neurology department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinical Medicine department, Miguel Hernández University, Elche, Spain
| | - Patricio Mas
- Pharmacy department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Sergio Reus
- Unit of Infectious Diseases, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinical Medicine department, Miguel Hernández University, Elche, Spain
| | - Rosario Sánchez-Martínez
- Internal Medicine department, Alicante General University Hospital - Alicante Institute of Sanitary and Biomedical Research (ISABIAL), Alicante, Spain
| | - Vicente Boix
- Unit of Infectious Diseases, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinical Medicine department, Miguel Hernández University, Elche, Spain
| | - Pablo Chico-Sánchez
- Preventive department, Alicante General University Hospital - - Alicante Institute of Sanitary and Biomedical Research (ISABIAL), Alicante, Spain
| | - José Sánchez-Payá
- Preventive department, Alicante General University Hospital - - Alicante Institute of Sanitary and Biomedical Research (ISABIAL), Alicante, Spain
| | - Joaquín Portilla
- Internal Medicine department, Alicante General University Hospital - Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinical Medicine department, Miguel Hernández University, Elche, Spain
| |
Collapse
|
28
|
Lüscher-Dias T, Siqueira Dalmolin RJ, de Paiva Amaral P, Alves TL, Schuch V, Franco GR, Nakaya HI. The evolution of knowledge on genes associated with human diseases. iScience 2022; 25:103610. [PMID: 35005554 PMCID: PMC8719018 DOI: 10.1016/j.isci.2021.103610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/05/2021] [Accepted: 12/08/2021] [Indexed: 12/15/2022] Open
Abstract
Thousands of biomedical scientific articles, including those describing genes associated with human diseases, are published every week. Computational methods such as text mining and machine learning algorithms are now able to automatically detect these associations. In this study, we used a cognitive computing text-mining application to construct a knowledge network comprising 3,723 genes and 99 diseases. We then tracked the yearly changes on these networks to analyze how our knowledge has evolved in the past 30 years. Our systems approach helped to unravel the molecular bases of diseases and detect shared mechanisms between clinically distinct diseases. It also revealed that multi-purpose therapeutic drugs target genes that are commonly associated with several psychiatric, inflammatory, or infectious disorders. By navigating this knowledge tsunami, we were able to extract relevant biological information and insights about human diseases.
Collapse
Affiliation(s)
- Thomaz Lüscher-Dias
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo Juliani Siqueira Dalmolin
- Bioinformatics Multidisciplinary Environment—BioME, IMD, Federal University of Rio Grande do Norte, Natal, RN, Brazil
- Department of Biochemistry, CB, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Tiago Lubiana Alves
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Viviane Schuch
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Glória Regina Franco
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Helder I. Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Scientific Platform Pasteur-University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
29
|
Khosravi M. Candidate Psychotropics against SARS - CoV - 2: A Narrative Review. PHARMACOPSYCHIATRY 2022; 55:16-23. [PMID: 34399430 DOI: 10.1055/a-1551-3756] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Since few therapeutic options are clinically accessible for coronavirus disease 2019 (COVID-19), effective, safe, and globally available pharmaceuticals need to be urgently developed to prevent severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) and alleviate the severity of COVID-19. In this regard, the present paper is intended to provide an extensive review of the clinical and preclinical evidence on the psychotropics' anti-SARS-CoV-2 effects, giving an insight into their potential applications for patients with a proven or high likelihood of COVID-19 pneumonia. The results showed that psychotropic drugs such as melatonin, lithium carbonate, valproate, olanzapine, quetiapine, clozapine, fluoxetine, escitalopram, fluvoxamine, and cannabidiol could help lower the mortality due to SARS-CoV-2 infection. According to these medications' direct immunomodulatory actions against the destructive cytokine storm, as well as other direct/indirect mechanisms (e. g., the endolysosomal pathway modulation, interactions with specific receptors, and membrane fusion), it was perceived that such drugs could effectively weaken the worsened immune response and avoid adult respiratory distress syndrome and acute lung injury. According to the author's analysis of the currently available evidence, there is significant support for psychotropics as complementary interventions during SARS-CoV-2 infection. However, further studies need to be carried out to assess the effects of the above psychotropic drugs in vitro and clinical settings.
Collapse
Affiliation(s)
- Mohsen Khosravi
- Department of Psychiatry and Clinical Psychology, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
30
|
Zannella C, Rinaldi L, Boccia G, Chianese A, Sasso FC, De Caro F, Franci G, Galdiero M. Regulation of m6A Methylation as a New Therapeutic Option against COVID-19. Pharmaceuticals (Basel) 2021; 14:ph14111135. [PMID: 34832917 PMCID: PMC8625908 DOI: 10.3390/ph14111135] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/23/2022] Open
Abstract
The rapid spread of SARS-CoV-2 and the resulting pandemic has led to a spasmodic search for approaches able to limit the diffusion of the disease. The epigenetic machinery has aroused considerable interest in the last decades, and much evidence has demonstrated that this type of modification could regulate the early stages of viral infection. Recently it was reported that N6-methyladenosine (m6A) influences SARS-CoV-2 replication, although its role remains to be further investigated. The knockdown of enzymes involved in the m6A pathway could represent an optimal strategy to deepen the epigenetic mechanism. In the present study, we blocked the catalytic activity of the fat mass and obesity-associated protein (FTO) by using the selective inhibitor rhein. We observed a strong broad-spectrum reduction of infectivity caused by various coronaviruses, including SARS-CoV-2. This effect could be due to the modulation of m6A levels and could allow identification of this modification as a new therapeutic target to treat SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Carla Zannella
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.Z.); (A.C.); (M.G.)
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.R.); (F.C.S.)
| | - Giovanni Boccia
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (G.B.); (F.D.C.)
| | - Annalisa Chianese
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.Z.); (A.C.); (M.G.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.R.); (F.C.S.)
| | - Francesco De Caro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (G.B.); (F.D.C.)
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (G.B.); (F.D.C.)
- Correspondence:
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.Z.); (A.C.); (M.G.)
| |
Collapse
|
31
|
Saiz ML, DeDiego ML, López-García D, Corte-Iglesias V, Baragaño Raneros A, Astola I, Asensi V, López-Larrea C, Suarez-Alvarez B. Epigenetic targeting of the ACE2 and NRP1 viral receptors limits SARS-CoV-2 infectivity. Clin Epigenetics 2021; 13:187. [PMID: 34635175 PMCID: PMC8504098 DOI: 10.1186/s13148-021-01168-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND SARS-CoV-2 uses the angiotensin-converting enzyme 2 (ACE2) and neuropilin-1 (NRP1) receptors for entry into cells, and the serine protease TMPRSS2 for S protein priming. Inhibition of protease activity or the engagement with ACE2 and NRP1 receptors has been shown to be an effective strategy for blocking infectivity and viral spreading. Valproic acid (VPA; 2-propylpentanoic acid) is an epigenetic drug approved for clinical use. It produces potent antiviral and anti-inflammatory effects through its function as a histone deacetylase (HDAC) inhibitor. Here, we propose VPA as a potential candidate to tackle COVID-19, in which rapid viral spread and replication, and hyperinflammation are crucial elements. RESULTS We used diverse cell lines (HK-2, Huh-7, HUVEC, Caco-2, and BEAS-2B) to analyze the effect of VPA and other HDAC inhibitors on the expression of the ACE-2 and NRP-1 receptors and their ability to inhibit infectivity, viral production, and the inflammatory response. Treatment with VPA significantly reduced expression of the ACE2 and NRP1 host proteins in all cell lines through a mechanism mediated by its HDAC inhibitory activity. The effect is maintained after SARS-CoV-2 infection. Consequently, the treatment of cells with VPA before infection impairs production of SARS-CoV-2 infectious viruses, but not that of other ACE2- and NRP1-independent viruses (VSV and HCoV-229E). Moreover, the addition of VPA 1 h post-infection with SARS-CoV-2 reduces the production of infectious viruses in a dose-dependent manner without significantly modifying the genomic and subgenomic messenger RNAs (gRNA and sg mRNAs) or protein levels of N protein. The production of inflammatory cytokines (TNF-α and IL-6) induced by TNF-α and SARS-CoV-2 infection is diminished in the presence of VPA. CONCLUSIONS Our data showed that VPA blocks three essential processes determining the severity of COVID-19. It downregulates the expression of ACE2 and NRP1, reducing the infectivity of SARS-CoV-2; it decreases viral yields, probably because it affects virus budding or virions stability; and it dampens the triggered inflammatory response. Thus, administering VPA could be considered a safe treatment for COVID-19 patients until vaccines have been rolled out across the world.
Collapse
Affiliation(s)
- Maria Laura Saiz
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Marta L DeDiego
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Darío López-García
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Viviana Corte-Iglesias
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Aroa Baragaño Raneros
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Ivan Astola
- Intensive Care Department, Hospital Universitario Central de Asturias, Oviedo, Spain.,Translational Microbiology Research Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Victor Asensi
- Infectious Diseases Unit, Translational Research in Infectious Diseases Group, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Carlos López-Larrea
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain. .,Department of Immunology, Hospital Universitario Central De Asturias, Oviedo, Spain.
| | - Beatriz Suarez-Alvarez
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
32
|
Singh D, Gupta S, Verma I, Morsy MA, Nair AB, Ahmed ASF. Hidden pharmacological activities of valproic acid: A new insight. Biomed Pharmacother 2021; 142:112021. [PMID: 34463268 DOI: 10.1016/j.biopha.2021.112021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/28/2021] [Accepted: 08/07/2021] [Indexed: 12/24/2022] Open
Abstract
Valproic acid (VPA) is an approved drug for managing epileptic seizures, bipolar disorders, and migraine. VPA has been shown to elevate the level of gamma-aminobutyric acid (GABA) in the brain through competitive inhibition of GABA transaminase, thus promoting the availability of synaptic GABA and facilitating GABA-mediated responses. VPA, which is a small chain of fatty acids, prevents histone deacetylases (HDACs). HDACs play a crucial role in chromatin remodeling and gene expression through posttranslational changes of chromatin-associated histones. Recent studies reported a possible effect of VPA against particular types of cancers. This effect was partially attributed to its role in regulating epigenetic modifications through the inhibition of HDACs, which affect the expression of genes associated with cell cycle control, cellular differentiation, and apoptosis. In this review, we summarize the current information on the actions of VPA in diseases such as diabetes mellitus, kidney disorders, neurodegenerative diseases, muscular dystrophy, and cardiovascular disorders.
Collapse
Affiliation(s)
- Dhirendra Singh
- Department of Pharmacology, M.M. College of Pharmacy, M.M. (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Sumeet Gupta
- Department of Pharmacology, M.M. College of Pharmacy, M.M. (Deemed to be University), Mullana, Ambala, Haryana, India.
| | - Inderjeet Verma
- Department of Pharmacology, M.M. College of Pharmacy, M.M. (Deemed to be University), Mullana, Ambala, Haryana, India
| | - Mohamed A Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
| | - Anroop B Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| |
Collapse
|
33
|
Angus-Leppan H, Guiloff AE, Benson K, Guiloff RJ. Navigating migraine care through the COVID-19 pandemic: an update. J Neurol 2021; 268:4388-4395. [PMID: 34002281 PMCID: PMC8128091 DOI: 10.1007/s00415-021-10610-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022]
Abstract
The worldwide treatment gap for migraine before COVID-19 inevitably widens as attention focuses on an international emergency. Migraine hits people particularly in their early and middle years, potentially reduces quality of life and productivity, and remains a common emergency presentation. This article examines the impact of COVID-19 on migraine, and changing aspects of migraine care during and after the pandemic. Many risk factors for severe COVID-19—older age, male gender, cardiac and respiratory diseases, diabetes, obesity, and immunosuppression—are less frequent in migraineurs. Telemedicine is effective for migraine follow-up, and needs ongoing evaluation. Most migraine treatments can start or continue in acute COVID-19, with care to avoid drug interactions. Close contact procedures (botulinum toxin, acupuncture and steroid injections) are avoided in lockdown or in the vulnerable. Secondary effects of COVID-19, including long COVID and its economic impact, are probably equal or greater in people with migraine. Migraine and other long-term conditions need adequate resourcing to prevent personal, social and economic suffering. Treating migraine, a sequel of COVID, potentially reduces the impact of long COVID.
Collapse
Affiliation(s)
- Heather Angus-Leppan
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
- Department of Clinical Neurosciences, Royal Free London NHS Foundation Trust, Pond Street, London, NW32QG, UK.
| | - Angelica E Guiloff
- Northern Medical Centre, Archway, London, UK
- Barts and the London School of Medicine and Dentistry, London, UK
| | | | - Roberto J Guiloff
- Faculty of Medicine, University of Chile, Santiago, Chile
- Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
34
|
Vicatos AI, Caira MR. Cyclodextrin complexes of the anticonvulsant agent valproic acid. CrystEngComm 2021. [DOI: 10.1039/d1ce01024g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Valproic acid (VAL) forms complexes with natural and derivatised cyclodextrins (CDs). Stoichiometries were deduced from NMR spectra, thermal and X-ray diffraction analyses, the latter revealing modes of VAL inclusion in CDs for the first time.
Collapse
Affiliation(s)
- A. I. Vicatos
- Centre for Supramolecular Chemistry Research, Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - M. R. Caira
- Centre for Supramolecular Chemistry Research, Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|