1
|
Blázquez AB, Mingo-Casas P, Quesada E, Priego EM, Pérez-Perez MJ, Martín-Acebes MA. Lipid-targeting antiviral strategies: Current state and future perspectives. Antiviral Res 2025; 236:106103. [PMID: 39947433 DOI: 10.1016/j.antiviral.2025.106103] [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: 12/11/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
There is an urgent need for antiviral compounds effective against currently known and future viral threats. The development of host-targeting antivirals (HTAs) appears as an alternative strategy to fight viral infections minimizing the potential of resistant mutant development and potentially leading to the identification of broad-spectrum antiviral agents. Among the host factors explored for HTA strategy, lipids constitute an attractive target as many viruses, even genetically diverse, hijack specific lipids during their lifecycle. Multiple repurposing efforts have been performed to analyze the antiviral properties of lipid-targeting compounds. These studies include the analysis of the effects of cholesterol lowering drugs such as statins, cholesterol transport inhibitors, sphingolipid modulators, de novo lipogenesis inhibitors blocking fatty acid synthesis, compounds targeting glycerophospholipids or drugs interfering with lipid droplet metabolism. This review is focused on the current status of lipid-based or lipid-targeting antiviral strategies and their potential for the development of antiviral therapies, with special emphasis on those studies that have reached advanced stages of development such as efficacy studies in animal models or clinical trials. Whereas there is still a long way to go, multiple proof-of-concept studies and clinical evidence reinforce the therapeutic potential of these strategies warranting their further development into effective antiviral therapies.
Collapse
Affiliation(s)
- Ana-Belén Blázquez
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain.
| | - Patricia Mingo-Casas
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain; Universidad Autónoma de Madrid (UAM, Escuela de Doctorado), Spain
| | | | | | | | - Miguel A Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain.
| |
Collapse
|
2
|
Kogachi L, Matozo T, Magalhães YT, Janoni Bayerlein M, Braga TC, Camargo FEC, da S. Souza KB, Forti FL, de Alencar BC. Myosin IXB protects immune cells from virus infection. J Gen Virol 2025; 106:002090. [PMID: 40167026 PMCID: PMC11962068 DOI: 10.1099/jgv.0.002090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Actin-associated proteins have been implicated in several stages of virus infection. However, the role of myosins, which are actin-dependent molecular motors, during virus infection and pathogenesis is poorly understood. Myosin IXB (Myo9b) is a member of the myosin family abundantly expressed in immune cells. Myo9b displays a RhoGTPase-activating protein domain capable of modulating actin dynamics by inhibiting RhoGTPase activity. To enquire upon Myo9b participation in virus infections, we have silenced Myo9b in U937 and Jurkat cells and infected them with vesicular stomatitis virus glycoprotein (VSV-G)-pseudotyped HIV-1. Myo9b-silenced U937 showed a remarkable increase of above ten times more HIV-VSV-G infection than control cells. We observed a similar pattern in Jurkat cell infection with both WT Env and VSV-G-pseudotyped HIV, albeit to a lesser extent. Myo9b-silenced U937 cells presented elevated levels of phosphorylated cofilin, but lower levels of polymerized actin. The use of a RhoA, B and C inhibitor, as well as a Rac1 inhibitor, reduced virus infection. Finally, we have also observed an increment in virus internalization and fusion in cells knockdown for Myo9b, which may explain the increase in virus infection. Taken together, our data suggests that Myo9b might hinder viral entry and infection by controlling the activity of RhoGTPases in immune cells.
Collapse
Affiliation(s)
- Leticia Kogachi
- Laboratory of Cell Biology of the Immune System, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Taís Matozo
- Laboratory of Cell Biology of the Immune System, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Yuli Thamires Magalhães
- Laboratory of Signaling in Biomolecular Systems, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Marina Janoni Bayerlein
- Laboratory of Cell Biology of the Immune System, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Tania Carolina Braga
- Laboratory of Cell Biology of the Immune System, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Felippe E. C. Camargo
- Laboratory of Cell Biology of the Immune System, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Kamilla B. da S. Souza
- Laboratory of Cell Biology of the Immune System, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Fábio Luis Forti
- Laboratory of Signaling in Biomolecular Systems, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Cunha de Alencar
- Laboratory of Cell Biology of the Immune System, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Neyrinck-Leglantier D, Tamagne M, Ben Rayana R, Many S, Pinheiro MK, Delorme AS, Andrieu M, Boilard E, Cognasse F, Hamzeh-Cognasse H, Perez-Patrigeon S, Lelievre JD, Pirenne F, Gallien S, Vingert B. Remodeling of immune system functions by extracellular vesicles. Front Immunol 2025; 16:1549107. [PMID: 40181981 PMCID: PMC11966064 DOI: 10.3389/fimmu.2025.1549107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/17/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The treatment of chronic viral infections can often bring viral replication under control. However, chronic immune activation persists and can lead to the development of comorbid conditions, such as cardiovascular disease and cancer. This is particularly true for people living with HIV (PLWH), who have significantly more extracellular vesicles from membrane budding, also called plasma microparticles (MPs), than healthy individuals (HDs), and a much more immunomodulatory phenotype. We hypothesized that the number and phenotypic heterogeneity of MPs can trigger a functional remodeling of immune responses in PLWH, preventing full immune restoration. Methods We investigated the rapid impact of three types of MPs - derived from membrane budding in platelets (CD41a+ PMPs), monocytes (CD14+ MMPs) and lymphocytes (CD3+ LMPs) in the plasma of PLWH or HDs-on four cell types (CD4+ and CD8+T lymphocytes, monocytes and DCs). Results These investigations of the short multiple interactions and functions of MPs with these cells revealed an increase in the secretion of cytokines such as IFNg, IL2, IL6, IL12, IL17 and TNFa by the immune cells studied following interactions with MPs. We show that this functional remodeling of immune cells depends not only on the number, but also on the phenotype of MPs. Conclusion These data suggest that the large numbers of MPs and their impact on functional remodeling in PLWH may be incompatible with the effective control of chronic infections, potentially leading to chronic immune activation and the onset of comorbid diseases.
Collapse
Affiliation(s)
- Deborah Neyrinck-Leglantier
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Marie Tamagne
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Raida Ben Rayana
- Service de Maladies Infectieuses et Immunologie Clinique, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Souganya Many
- Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique (CNRS) UMR8104, Université Paris-Cité, Paris, France
| | - Marion Klea Pinheiro
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Adèle Silane Delorme
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Muriel Andrieu
- Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique (CNRS) UMR8104, Université Paris-Cité, Paris, France
| | - Eric Boilard
- Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France
- Univ Jean Monnet, Mines Saint-Étienne, INSERM, U1059 Sainbiose, Saint-Étienne, France
| | - Hind Hamzeh-Cognasse
- Univ Jean Monnet, Mines Saint-Étienne, INSERM, U1059 Sainbiose, Saint-Étienne, France
| | | | - Jean-Daniel Lelievre
- Service de Maladies Infectieuses et Immunologie Clinique, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - France Pirenne
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| | - Sébastien Gallien
- Service de Maladies Infectieuses et Immunologie Clinique, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Benoît Vingert
- Univ Paris Est-Creteil (UPEC), Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Mondor de la Recherche Biomédicale (IMRB), Creteil, France
- Etablissement Français du Sang (EFS), Ivry-sur-Seine, France
- Laboratory of Excellence, Biogénèse et Pathologies du Globule Rouge (GR-Ex), Paris, France
| |
Collapse
|
4
|
Kelly SH, Nightingale S, Gupta RK, Collier DA. HIV Cerebrospinal Fluid Escape: Interventions for the Management, Current Evidence and Future Perspectives. Trop Med Infect Dis 2025; 10:45. [PMID: 39998049 PMCID: PMC11860496 DOI: 10.3390/tropicalmed10020045] [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: 11/19/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Neurocognitive impairment is an important cause of HIV-associated morbidity. The advent of antiretroviral therapy (ART) has shifted the spectrum of HIV-associated cognitive impairment from HIV-associated dementia to milder forms of cognitive impairment. Independent replication of HIV within the central nervous system in those on effective ART with peripheral suppression is a recognised phenomenon known as cerebrospinal fluid (CSF) HIV RNA escape. CSF HIV RNA escape is independently associated with neurocognitive impairment but has also been detected in asymptomatic persons with HIV. The current consensus for management of CSF HIV RNA escape is based on expert opinion rather than empirical evidence. The current evidence suggests having a low threshold to investigate for CSF HIV RNA escape and optimising ART based on resistance profiles. The use of central nervous system (CNS) penetration effectiveness scores is no longer recommended. The evidence for statins, SSRIs, minocycline, lithium and valproate is limited to small-scale studies. There are potential new developments in the form of nanoparticles, Janus Kinase inhibitors and latency reversal agents.
Collapse
Affiliation(s)
- Sophie H. Kelly
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK;
- Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK
| | - Sam Nightingale
- Neuroscience Institute, University of Cape Town, Cape Town 7700, South Africa;
| | - Ravindra K. Gupta
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK;
- Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Africa Health Research Institute, Durban 4013, South Africa
| | - Dami A. Collier
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK;
- Department of Pathology, University of Cambridge, Cambridge CB2 1TN, UK
| |
Collapse
|
5
|
Abavisani M, Hoseinzadeh M, Khayami R, Kodori M, Soleimanpour S, Sahebkar A. Statins, Allies against Antibiotic Resistance? Curr Med Chem 2025; 32:729-752. [PMID: 37644745 DOI: 10.2174/0929867331666230829141301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/22/2023] [Accepted: 07/20/2023] [Indexed: 08/31/2023]
Abstract
Due to the ever-increasing rate of antibacterial resistance, the search for effective antibacterial agents has become imperative. Researchers have investigated the potential antimicrobial properties of various classes of nonantibiotic drugs. Statins are a group of antihyperlipidemic drugs with several cholesterol-independent effects, including antiinflammatory, immune-modulating, antioxidant, and antibacterial effects. In vitro and in vivo studies have demonstrated the antibacterial properties of statins against various grampositive and gram-negative bacteria. Simvastatin and atorvastatin are the most potent members of the family. Their antibacterial effect can be attributed to several direct and indirect mechanisms. Bacterial invasion, growth, and virulence are affected by statins. However, since in vitro minimum inhibitory concentrations (MICs) are significantly higher than serum concentrations at the lipid-lowering dosage, indirect mechanisms have been suggested to explain the positive clinical results, including reducing inflammation and improving immune response capacity. Further, statins have shown promising results when combined with antibiotics and other antibacterial agents, such as triazenes and silver nanoparticles. Despite this, the controversial aspects of statins have cast doubt on their efficacy as a possible solution for antibacterial resistance, and further research is required. Consequently, this review will examine in detail the current clinical and in vitro findings and controversies regarding statins' antibacterial properties and their relevance to antibacterial resistance.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Melika Hoseinzadeh
- Dental Research Center, Mashhad Dental School, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Khayami
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mansoor Kodori
- Non-communicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Saman Soleimanpour
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Wang Y, Gao L. Cholesterol: A friend to viruses. Int Rev Immunol 2024; 43:248-262. [PMID: 38372266 DOI: 10.1080/08830185.2024.2314577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/21/2023] [Accepted: 01/28/2024] [Indexed: 02/20/2024]
Abstract
Cholesterol is a key life-sustaining molecule which regulates membrane fluidity and serves as a signaling mediator. Cholesterol homeostasis is closely related to various pathological conditions including tumor, obesity, atherosclerosis, Alzheimer's disease and viral infection. Viral infection disrupts host cholesterol homeostasis, facilitating their own survival. Meanwhile, the host cells strive to reduce cholesterol accessibility to limit viral infection. This review focuses on the regulation of cholesterol metabolism and the role of cholesterol in viral infection, specifically providing an overview of cholesterol as a friend to promote viral entry, replication, assembly, release and immune evasion, which might inspire valuable thinking for pathogenesis and intervention of viral infection.
Collapse
Affiliation(s)
- Yingchun Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
7
|
Yu C, Wang G, Liu Q, Zhai J, Xue M, Li Q, Xian Y, Zheng C. Host antiviral factors hijack furin to block SARS-CoV-2, ebola virus, and HIV-1 glycoproteins cleavage. Emerg Microbes Infect 2023; 12:2164742. [PMID: 36591809 PMCID: PMC9897805 DOI: 10.1080/22221751.2022.2164742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Viral envelope glycoproteins are crucial for viral infections. In the process of enveloped viruses budding and release from the producer cells, viral envelope glycoproteins are presented on the viral membrane surface as spikes, promoting the virus's next-round infection of target cells. However, the host cells evolve counteracting mechanisms in the long-term virus-host co-evolutionary processes. For instance, the host cell antiviral factors could potently suppress viral replication by targeting their envelope glycoproteins through multiple channels, including their intracellular synthesis, glycosylation modification, assembly into virions, and binding to target cell receptors. Recently, a group of studies discovered that some host antiviral proteins specifically recognized host proprotein convertase (PC) furin and blocked its cleavage of viral envelope glycoproteins, thus impairing viral infectivity. Here, in this review, we briefly summarize several such host antiviral factors and analyze their roles in reducing furin cleavage of viral envelope glycoproteins, aiming at providing insights for future antiviral studies.
Collapse
Affiliation(s)
- Changqing Yu
- School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, People’s Republic of China
| | - Guosheng Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Qiang Liu
- Nanchong Key Laboratory of Disease Prevention, Control and Detection in Livestock and Poultry, Nanchong Vocational and Technical College, Nanchong, People’s Republic of China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, People’s Republic of China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China,Mengzhou Xue
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China,Qiang Li
| | - Yuanhua Xian
- School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, People’s Republic of China,Yuanhua Xian
| | - Chunfu Zheng
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China,Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Canada, Chunfu Zheng
| |
Collapse
|
8
|
Tewari DN, Biswas A, Chakrabarti AK, Dutta S. AMFR promotes innate immunity activation and proteasomal degradation of HMGCR in response to influenza virus infection in A549 cells. Virology 2023; 587:109875. [PMID: 37703797 DOI: 10.1016/j.virol.2023.109875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/15/2023]
Abstract
Differential regulation of the 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR), which is considered the rate-limiting enzyme of the cholesterol biosynthesis pathway, has been reported in case of infection with many viruses. In our study, we have found that influenza virus infection decreases total cellular cholesterol level which is directly related to the downregulation of HMGCR protein. We found that HMGCR is degraded through ubiquitination and proteasomal-mediated pathway upon viral infection. Upregulation of Autocrine Motility Factor Receptor (AMFR), which is an E3-ubiquitin ligase of HMGCR, was also observed. Furthermore, knockdown of AMFR inhibits ubiquitination of HMGCR and also leads to inactivation of the innate immunity components TANK-binding kinase 1 (TBK1) and Interferon regulatory factor 3 (IRF3). Our study is the first to show the role of HMGCR and AMFR in influenza virus infection and reveals that AMFR plays a crucial role in the downregulation of HMGCR and the activation of innate immunity following influenza virus infection.
Collapse
Affiliation(s)
- Devendra Nath Tewari
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010, India
| | - Asim Biswas
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010, India
| | - Alok Kumar Chakrabarti
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010, India.
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010, India
| |
Collapse
|
9
|
Sanz M, Weideman AMK, Ward AR, Clohosey ML, Garcia-Recio S, Selitsky SR, Mann BT, Iannone MA, Whitworth CP, Chitrakar A, Garrido C, Kirchherr J, Coffey AR, Tsai YH, Samir S, Xu Y, Copertino D, Bosque A, Jones BR, Parker JS, Hudgens MG, Goonetilleke N, Soriano-Sarabia N. Aminobisphosphonates reactivate the latent reservoir in people living with HIV-1. Front Immunol 2023; 14:1219250. [PMID: 37744358 PMCID: PMC10516574 DOI: 10.3389/fimmu.2023.1219250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Antiretroviral therapy (ART) is not curative due to the existence of cellular reservoirs of latent HIV-1 that persist during therapy. Current research efforts to cure HIV-1 infection include "shock and kill" strategies to disrupt latency using small molecules or latency-reversing agents (LRAs) to induce expression of HIV-1 enabling cytotoxic immune cells to eliminate infected cells. The modest success of current LRAs urges the field to identify novel drugs with increased clinical efficacy. Aminobisphosphonates (N-BPs) that include pamidronate, zoledronate, or alendronate, are the first-line treatment of bone-related diseases including osteoporosis and bone malignancies. Here, we show the use of N-BPs as a novel class of LRA: we found in ex vivo assays using primary cells from ART-suppressed people living with HIV-1 that N-BPs induce HIV-1 from latency to levels that are comparable to the T cell activator phytohemagglutinin (PHA). RNA sequencing and mechanistic data suggested that reactivation may occur through activation of the activator protein 1 signaling pathway. Stored samples from a prior clinical trial aimed at analyzing the effect of alendronate on bone mineral density, provided further evidence of alendronate-mediated latency reversal and activation of immune effector cells. Decay of the reservoir measured by IPDA was however not detected. Our results demonstrate the novel use of N-BPs to reverse HIV-1 latency while inducing immune effector functions. This preliminary evidence merits further investigation in a controlled clinical setting possibly in combination with therapeutic vaccination.
Collapse
Affiliation(s)
- Marta Sanz
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington, DC, United States
| | - Ann Marie K. Weideman
- Biostatistics Core, Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Adam R. Ward
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington, DC, United States
- Department of Infectious Diseases, Weill Cornell Medicine, New York, NY, United States
| | - Matthew L. Clohosey
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susana Garcia-Recio
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sara R. Selitsky
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Brendan T. Mann
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington, DC, United States
| | - Marie Anne Iannone
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chloe P. Whitworth
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alisha Chitrakar
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington, DC, United States
| | - Carolina Garrido
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jennifer Kirchherr
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alisha R. Coffey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yi- Hsuan Tsai
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Shahryar Samir
- Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yinyan Xu
- Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dennis Copertino
- Department of Infectious Diseases, Weill Cornell Medicine, New York, NY, United States
| | - Alberto Bosque
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington, DC, United States
| | - Brad R. Jones
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington, DC, United States
- Department of Infectious Diseases, Weill Cornell Medicine, New York, NY, United States
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael G. Hudgens
- Biostatistics Core, Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nilu Goonetilleke
- Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Natalia Soriano-Sarabia
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington, DC, United States
| |
Collapse
|
10
|
Abstract
There is a large global unmet need for the development of countermeasures to combat hundreds of viruses known to cause human disease and for the establishment of a therapeutic portfolio for future pandemic preparedness. Most approved antiviral therapeutics target proteins encoded by a single virus, providing a narrow spectrum of coverage. This, combined with the slow pace and high cost of drug development, limits the scalability of this direct-acting antiviral (DAA) approach. Here, we summarize progress and challenges in the development of broad-spectrum antivirals that target either viral elements (proteins, genome structures, and lipid envelopes) or cellular proviral factors co-opted by multiple viruses via newly discovered compounds or repurposing of approved drugs. These strategies offer new means for developing therapeutics against both existing and emerging viral threats that complement DAAs.
Collapse
Affiliation(s)
- Marwah Karim
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, and
| | - Chieh-Wen Lo
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, and
| | - Shirit Einav
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA
| |
Collapse
|
11
|
Innocenti M. Investigating Mammalian Formins with SMIFH2 Fifteen Years in: Novel Targets and Unexpected Biology. Int J Mol Sci 2023; 24:ijms24109058. [PMID: 37240404 DOI: 10.3390/ijms24109058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The mammalian formin family comprises fifteen multi-domain proteins that regulate actin dynamics and microtubules in vitro and in cells. Evolutionarily conserved formin homology (FH) 1 and 2 domains allow formins to locally modulate the cell cytoskeleton. Formins are involved in several developmental and homeostatic processes, as well as human diseases. However, functional redundancy has long hampered studies of individual formins with genetic loss-of-function approaches and prevents the rapid inhibition of formin activities in cells. The discovery of small molecule inhibitor of formin homology 2 domains (SMIFH2) in 2009 was a disruptive change that provided a powerful chemical tool to explore formins' functions across biological scales. Here, I critically discuss the characterization of SMIFH2 as a pan-formin inhibitor, as well as growing evidence of unexpected off-target effects. By collating the literature and information hidden in public repositories, outstanding controversies and fundamental open questions about the substrates and mechanism of action of SMIFH2 emerge. Whenever possible, I propose explanations for these discrepancies and roadmaps to address the paramount open questions. Furthermore, I suggest that SMIFH2 be reclassified as a multi-target inhibitor for its appealing activities on proteins involved in pathological formin-dependent processes. Notwithstanding all drawbacks and limitations, SMIFH2 will continue to prove useful in studying formins in health and disease in the years to come.
Collapse
Affiliation(s)
- Metello Innocenti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| |
Collapse
|
12
|
Gee YJ, Sea YL, Lal SK. Viral modulation of lipid rafts and their potential as putative antiviral targets. Rev Med Virol 2023; 33:e2413. [PMID: 36504273 DOI: 10.1002/rmv.2413] [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: 06/27/2022] [Revised: 11/12/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022]
Abstract
Lipid rafts are ubiquitous in cells. They are identified as cholesterol and glycosphingolipid enriched microdomains on cellular membranes. They serve as platforms for cellular communications by functioning in signal transduction and membrane trafficking. Such structural organisation fulfils cellular needs for normal function, but at the same time increases vulnerability of cells to pathogen invasion. Viruses rely heavily on lipid rafts in basically every stage of the viral life cycle for successful infection. Various mechanisms of lipid rafts modification exploited by diverse viruses for attachment, internalisation, membrane fusion, genome replication, assembly and release have been brought to light. This review focuses on virus-raft interactions and how a wide range of viruses manipulate lipid rafts at distinct stages of infection. The importance of virus-raft interactions in viral infections has inspired researchers to discover and develop antivirals that target this interaction, such as statins, methyl-β-cyclodextrin, viperin, 25-hydroxycholesterol and even anti-malarial drugs. The therapeutic modulations of lipid rafts as potential antiviral intervention from in vitro and in vivo evidence are discussed herein.
Collapse
Affiliation(s)
- Yee Jing Gee
- School of Science, Monash University, Bandar Sunway, Selangor DE, Malaysia
| | - Yi Lin Sea
- School of Science, Monash University, Bandar Sunway, Selangor DE, Malaysia
| | - Sunil Kumar Lal
- School of Science, Monash University, Bandar Sunway, Selangor DE, Malaysia.,Tropical Medicine & Biology Platform, Monash University, Bandar Sunway, Selangor DE, Malaysia
| |
Collapse
|
13
|
Sanz M, Weideman AMK, Ward AR, Clohosey ML, Garcia-Recio S, Selitsky SR, Mann BT, Iannone MA, Whitworth CP, Chitrakar A, Garrido C, Kirchherr J, Coffey AR, Tsai YH, Samir S, Xu Y, Copertino D, Bosque A, Jones BR, Parker JS, Hudgens MG, Goonetilleke N, Soriano-Sarabia N. Aminobisphosphonates reactivate the latent reservoir in people living with HIV-1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527421. [PMID: 36798291 PMCID: PMC9934553 DOI: 10.1101/2023.02.07.527421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Antiretroviral therapy (ART) is not curative due to the existence of cellular reservoirs of latent HIV-1 that persist during therapy. Current research efforts to cure HIV-1 infection include "shock and kill" strategies to disrupt latency using small molecules or latency-reversing agents (LRAs) to induce expression of HIV-1 enabling cytotoxic immune cells to eliminate infected cells. The modest success of current LRAs urges the field to identify novel drugs with increased clinical efficacy. Aminobisphosphonates (N-BPs) that include pamidronate, zoledronate, or alendronate, are the first-line treatment of bone-related diseases including osteoporosis and bone malignancies. Here, we show the use of N-BPs as a novel class of LRA: we found in ex vivo assays using primary cells from ART-suppressed people living with HIV-1 that N-BPs induce HIV-1 from latency to levels that are comparable to the T cell activator phytohemagglutinin (PHA). RNA sequencing and mechanistic data suggested that reactivation may occur through activation of the activator protein 1 signaling pathway. Stored samples from a prior clinical trial aimed at analyzing the effect of alendronate on bone mineral density, provided further evidence of alendronate-mediated latency reversal and activation of immune effector cells. Decay of the reservoir measured by IPDA was however not detected. Our results demonstrate the novel use of N-BPs to reverse HIV-1 latency while inducing immune effector functions. This preliminary evidence merits further investigation in a controlled clinical setting possibly in combination with therapeutic vaccination.
Collapse
Affiliation(s)
- Marta Sanz
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington DC, USA
| | - Ann Marie K. Weideman
- Department of Biostatistics, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Adam R. Ward
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington DC, USA
- Department of Infectious Diseases, Weill Cornell Medicine, New York, USA
| | - Matthew L. Clohosey
- UNC HIV-1 Cure Center, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Susana Garcia-Recio
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Sara R. Selitsky
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Brendan T. Mann
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington DC, USA
| | - Marie Anne Iannone
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Chloe P. Whitworth
- UNC HIV-1 Cure Center, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Alisha Chitrakar
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington DC, USA
| | - Carolina Garrido
- UNC HIV-1 Cure Center, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Jennifer Kirchherr
- UNC HIV-1 Cure Center, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Alisha R. Coffey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Yi-Hsuan Tsai
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Shahryar Samir
- Microbiology & Immunology, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Yinyan Xu
- Microbiology & Immunology, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Dennis Copertino
- Department of Infectious Diseases, Weill Cornell Medicine, New York, USA
| | - Alberto Bosque
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington DC, USA
| | - Brad R. Jones
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington DC, USA
- Department of Infectious Diseases, Weill Cornell Medicine, New York, USA
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Michael G. Hudgens
- Department of Biostatistics, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Nilu Goonetilleke
- Microbiology & Immunology, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Natalia Soriano-Sarabia
- Department of Microbiology Immunology and Tropical Medicine, the George Washington University, Washington DC, USA
| |
Collapse
|
14
|
Mustafin R. Prospects for the use of statins in antiviral therapy. CLINICAL MICROBIOLOGY AND ANTIMICROBIAL CHEMOTHERAPY 2023; 25:56-67. [DOI: 10.36488/cmac.2023.1.56-67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Inhibitors of hydroxymethylglutaryl-CoA reductase, in addition to suppressing cholesterol synthesis, have an antiviral effect. Clinical studies have shown antiviral efficacy of statins against COVID-19, HCV, HBV, RSV, HIV, influenza viruses. The ability of statins to inhibit influenza viruses, COVID-19, RSV, HIV, as well as Ebola, Zika, Dengue, Coxsackie, rotaviruses, ADV, HDV, HHV was experimentally confirmed. Statins can also enhance the effects of antiviral drugs, making them more effective in treating infections. Therefore, the use of statins in the complex therapy of viral infections is promising. In addition, the role of influenza viruses, T-cell leukemia and herpesviruses, HIV, HBV, HCV, HPV in the development of atherosclerosis has been identified, so the use of statins in complex treatment is also necessary to correct endothelial dysfunction that occurs under the influence of viruses. Since the activity of retroelements that are evolutionarily related to exogenous viruses increases with aging, it has been suggested that retrotransposons can also be targets for statins. This is evidenced by a change in the expression of non-coding RNAs under the action of statins, since the key sources of non-coding RNAs are retroelements. This property may be an additional factor in the prescription of statins to increase life expectancy, in addition to the prevention and treatment of atherosclerosis, since pathological activation of retroelements are the causes of aging. Viruses, like retroelements, are involved in the pathogenesis of malignant neoplasms, in the treatment of which statins have shown their effectiveness and the ability to enhance the effect of anticancer drugs, overcoming chemoresistance (similar to the potentiation of antiviral drugs). One of the mechanisms of this activity of statins may be their effect on retroelements and viruses.
Collapse
|
15
|
Fitch KV, Fulda ES, Grinspoon SK. Statins for primary cardiovascular disease prevention among people with HIV: emergent directions. Curr Opin HIV AIDS 2022; 17:293-300. [PMID: 35938463 PMCID: PMC9415230 DOI: 10.1097/coh.0000000000000752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW While people with HIV (PWH) are living longer due to advances in antiretroviral therapy, recent data have demonstrated an increased risk of cardiovascular disease (CVD) among this population. This increased risk is thought to be due to both traditional (for example, smoking, diabetes) and HIV-specific (for example, inflammation, persistent immune activation) risk factors. This review focuses on the potential for statin therapy to mitigate this increased risk. RECENT FINDINGS Several randomized clinical trials have demonstrated that statins, a class of lipid-lowering medications, are effective as a primary CVD prevention strategy among people without HIV. Among PWH, statins have been shown to lower cholesterol, exert immunomodulatory effects, stabilize coronary atherosclerotic plaque, and even induce plaque regression. SUMMARY Prevention of CVD among the aging population of people with controlled, but chronic, HIV is vital. Data exploring primary prevention in this context are thus far limited. The Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) is ongoing; this trial will inform the field by investigating the effects of pitavastatin calcium as a primary prevention strategy for major adverse cardiovascular events among PWH on antiretroviral therapy (ART) at low-to-moderate traditional CVD risk.
Collapse
Affiliation(s)
- Kathleen V Fitch
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
16
|
Atorvastatin Rapidly Reduces Hepatitis B Viral Load in Combination with Tenofovir: A Prospective Clinical Trial. CANADIAN JOURNAL OF INFECTIOUS DISEASES AND MEDICAL MICROBIOLOGY 2022; 2022:3443813. [PMID: 35873362 PMCID: PMC9303483 DOI: 10.1155/2022/3443813] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/14/2022] [Accepted: 06/24/2022] [Indexed: 11/18/2022]
Abstract
Objective and Aim. Atorvastatin inhibits cholesterol synthesis which is critically important in the formation of the viral envelope and secretion. The efficacy and safety of giving atorvastatin (40 mg/day) as an adjunct to tenofovir in the treatment of hepatitis B (HBV) were assessed. Method. In this single-blind clinical trial, 40 patients with active chronic hepatitis B were randomly allocated to treatment or control groups. The treatment group received the standard treatment for chronic HBV (300 mg tenofovir twice a day) along with 40 mg/day atorvastatin for 12 months, while the control group received a placebo once daily in addition to the standard tenofovir regimen. Serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and HBV DNA copy numbers were measured at the beginning of the treatment and 1, 3, 6, 9, 12 months later. Results. One month after starting the treatment, the HBV copy number in the atorvastatin + tenofovir-treated group was significantly lower, by 200×, compared with the control group. After three months of the treatment, there was no detectable HBV DNA in 50% of the atorvastatin + tenofovir-treated group compared with 30% in the control group. The half-life of plasma viral load was 2.03 and 3.32 months in the atorvastatin + tenofovir-treated and control groups, respectively. No adverse events due to taking atorvastatin were observed. Conclusions. The combination of atorvastatin with tenofovir increased antiviral activity and led to a faster recovery from viral infection. Therefore, this modality can be recommended as a safe combination therapy for chronic hepatitis B patients.
Collapse
|
17
|
Barrantes FJ. The constellation of cholesterol-dependent processes associated with SARS-CoV-2 infection. Prog Lipid Res 2022; 87:101166. [PMID: 35513161 PMCID: PMC9059347 DOI: 10.1016/j.plipres.2022.101166] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 01/11/2023]
Abstract
The role of cholesterol in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other coronavirus-host cell interactions is currently being discussed in the context of two main scenarios: i) the presence of the neutral lipid in cholesterol-rich lipid domains involved in different steps of the viral infection and ii) the alteration of metabolic pathways by the virus over the course of infection. Cholesterol-enriched lipid domains have been reported to occur in the lipid envelope membrane of the virus, in the host-cell plasma membrane, as well as in endosomal and other intracellular membrane cellular compartments. These membrane subdomains, whose chemical and physical properties distinguish them from the bulk lipid bilayer, have been purported to participate in diverse phenomena, from virus-host cell fusion to intracellular trafficking and exit of the virions from the infected cell. SARS-CoV-2 recruits many key proteins that participate under physiological conditions in cholesterol and lipid metabolism in general. This review analyses the status of cholesterol and lipidome proteins in SARS-CoV-2 infection and the new horizons they open for therapeutic intervention.
Collapse
Affiliation(s)
- Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Institute for Biomedical Research (BIOMED), Faculty of Medical Sciences, UCA-CONICET, Av. Alicia Moreau de Justo 1600, C1107AFF Buenos Aires, Argentina.
| |
Collapse
|
18
|
Liu C, Yan W, Shi J, Wang S, Peng A, Chen Y, Huang K. Biological Actions, Implications, and Cautions of Statins Therapy in COVID-19. Front Nutr 2022; 9:927092. [PMID: 35811982 PMCID: PMC9257176 DOI: 10.3389/fnut.2022.927092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) showed worse prognosis and higher mortality in individuals with obesity. Dyslipidemia is a major link between obesity and COVID-19 severity. Statins as the most common lipid regulating drugs have shown favorable effects in various pathophysiological states. Importantly, accumulating observational studies have suggested that statin use is associated with reduced risk of progressing to severe illness and in-hospital death in COVID-19 patients. Possible explanations underlie these protective impacts include their abilities of reducing cholesterol, suppressing viral entry and replication, anti-inflammation and immunomodulatory effects, as well as anti-thrombosis and anti-oxidative properties. Despite these benefits, statin therapies have side effects that should be considered, such as elevated creatinine kinase, liver enzyme and serum glucose levels, which are already elevated in severe COVID-19. Concerns are also raised whether statins interfere with the efficacy of COVID-19 vaccines. Randomized controlled trials are being conducted worldwide to confirm the values of statin use for COVID-19 treatment. Generally, the results suggest no necessity to discontinue statin use, and no evidence suggesting interference between statins and COVID-19 vaccines. However, concomitant administration of statins and COVID-19 antiviral drug Paxlovid may increase statin exposure and the risk of adverse effects, because most statins are metabolized mainly through CYP3A4 which is potently inhibited by ritonavir, a major component of Paxlovid. Therefore, more clinical/preclinical studies are still warranted to understand the benefits, harms and mechanisms of statin use in the context of COVID-19.
Collapse
Affiliation(s)
- Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyao Yan
- Department of Pharmacy, Wuhan Fourth Hospital, Wuhan, China
| | - Jiajian Shi
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shun Wang
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anlin Peng
- Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Tongji-Rongcheng Center for Biomedicine, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Ha CHX, Lee NK, Rahman T, Hwang SS, Yam WK, Chee XW. Repurposing FDA-approved drugs as HIV-1 integrase inhibitors: an in silico investigation. J Biomol Struct Dyn 2022; 41:2146-2159. [PMID: 35067186 DOI: 10.1080/07391102.2022.2028677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Human Immunodeficiency Virus (HIV) infection is a global pandemic that has claimed 33 million lives to-date. One of the most efficacious treatments for naïve or pretreated HIV patients is the HIV integrase strand transfer inhibitors (INSTIs). However, given that HIV treatment is life-long, the emergence of HIV strains resistant to INSTIs is an imminent challenge. In this work, we showed two best regression QSAR models that were constructed using a boosted Random Forest algorithm (r2 = 0.998, q210CV = 0.721, q2external_test = 0.754) and a boosted K* algorithm (r2 = 0.987, q210CV = 0.721, q2external_test = 0.758) to predict the pIC50 values of INSTIs. Subsequently, the regression QSAR models were deployed against the Drugbank database for drug repositioning. The top-ranked compounds were further evaluated for their target engagement activity using molecular docking studies and accelerated Molecular Dynamics simulation. Lastly, their potential as INSTIs were also evaluated from our literature search. Our study offers the first example of a large-scale regression QSAR modelling effort for discovering highly active INSTIs to combat HIV infection.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Christopher Heng Xuan Ha
- Faculty of Engineering, Computing and Science, Swinburne University of Technology, Sarawak, Malaysia
| | - Nung Kion Lee
- Faculty of Cognitive Sciences and Human Development, Universiti Malaysia Sarawak, Sarawak, Malaysia
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Siaw San Hwang
- Faculty of Engineering, Computing and Science, Swinburne University of Technology, Sarawak, Malaysia
| | - Wai Keat Yam
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Kuala Lumpur, Malaysia
| | - Xavier Wezen Chee
- Faculty of Engineering, Computing and Science, Swinburne University of Technology, Sarawak, Malaysia
| |
Collapse
|
20
|
Immunotherapy with Cell-Based Biological Drugs to Cure HIV-1 Infection. Cells 2021; 11:cells11010077. [PMID: 35011639 PMCID: PMC8750418 DOI: 10.3390/cells11010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/21/2021] [Accepted: 12/25/2021] [Indexed: 11/17/2022] Open
Abstract
Since its discovery 35 years ago, there have been no therapeutic interventions shown to enable full HIV-1 remission. Combined antiretroviral therapy (cART) has achieved the sustained control of HIV-1 replication, however, the life-long treatment does not eradicate long-lived latently infected reservoirs and can result in multiple side effects including the development of multidrug-resistant escape mutants. Antibody-based treatments have emerged as alternative approaches for a HIV-1 cure. Here, we will review clinical advances in coreceptor-targeting antibodies, with respect to anti-CCR5 antibodies in particular, which are currently being generated to target the early stages of infection. Among the Env-specific antibodies widely accepted as relevant in cure strategies, the potential role of those targeting CD4-induced (CD4i) epitopes of the CD4-binding site (CD4bs) in eliminating HIV-1 infected cells has gained increasing interest and will be presented. Together, with approaches targeting the HIV-1 replication cycle, we will discuss the strategies aimed at boosting and modulating specific HIV-1 immune responses, highlighting the harnessing of TLR agonists for their dual role as latency reverting agents (LRAs) and immune-modulatory compounds. The synergistic combinations of different approaches have shown promising results to ultimately enable a HIV-1 cure.
Collapse
|
21
|
Zapatero-Belinchón FJ, Moeller R, Lasswitz L, van Ham M, Becker M, Brogden G, Rosendal E, Bi W, Carriquí-Madroñal B, Islam K, Lenman A, Gunesch AP, Kirui J, Pietschmann T, Överby AK, Jänsch L, Gerold G. Fluvastatin mitigates SARS-CoV-2 infection in human lung cells. iScience 2021; 24:103469. [PMID: 34812415 PMCID: PMC8599137 DOI: 10.1016/j.isci.2021.103469] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/08/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Clinical data of patients suffering from COVID-19 indicates that statin therapy, used to treat hypercholesterolemia, is associated with a better disease outcome. Whether statins directly affect virus replication or influence the clinical outcome through modulation of immune responses is unknown. We therefore investigated the effect of statins on SARS-CoV-2 infection in human lung cells and found that only fluvastatin inhibited low and high pathogenic coronaviruses in vitro and ex vivo in a dose-dependent manner. Quantitative proteomics revealed that fluvastatin and other tested statins modulated the cholesterol synthesis pathway without altering innate antiviral immune responses in infected lung epithelial cells. However, fluvastatin treatment specifically downregulated proteins that modulate protein translation and viral replication. Collectively, these results support the notion that statin therapy poses no additional risk to individuals exposed to SARS-CoV-2 and that fluvastatin has a moderate beneficial effect on SARS-CoV-2 infection of human lung cells.
Collapse
Affiliation(s)
- Francisco J. Zapatero-Belinchón
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Clinical Microbiology, Virology, Umeå University, 90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90185 Umeå, Sweden
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Rebecca Moeller
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Lisa Lasswitz
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Marco van Ham
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Miriam Becker
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Clinical Microbiology, Virology, Umeå University, 90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90185 Umeå, Sweden
| | - Graham Brogden
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Ebba Rosendal
- Department of Clinical Microbiology, Virology, Umeå University, 90185 Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), R893+F4 Umeå, Sweden
| | - Wenjie Bi
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Belén Carriquí-Madroñal
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Koushikul Islam
- Department of Clinical Microbiology, Virology, Umeå University, 90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90185 Umeå, Sweden
| | - Annasara Lenman
- Department of Clinical Microbiology, Virology, Umeå University, 90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90185 Umeå, Sweden
| | - Antonia P. Gunesch
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, 30625 Hannover, Germany
| | - Jared Kirui
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Thomas Pietschmann
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, 30625 Hannover, Germany
| | - Anna K. Överby
- Department of Clinical Microbiology, Virology, Umeå University, 90185 Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), R893+F4 Umeå, Sweden
| | - Lothar Jänsch
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Gisa Gerold
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, 30625 Hannover, Germany
- Department of Clinical Microbiology, Virology, Umeå University, 90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90185 Umeå, Sweden
- Department of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| |
Collapse
|
22
|
Palacios-Rápalo SN, De Jesús-González LA, Cordero-Rivera CD, Farfan-Morales CN, Osuna-Ramos JF, Martínez-Mier G, Quistián-Galván J, Muñoz-Pérez A, Bernal-Dolores V, del Ángel RM, Reyes-Ruiz JM. Cholesterol-Rich Lipid Rafts as Platforms for SARS-CoV-2 Entry. Front Immunol 2021; 12:796855. [PMID: 34975904 PMCID: PMC8719300 DOI: 10.3389/fimmu.2021.796855] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Since its appearance, the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2), the causal agent of Coronavirus Disease 2019 (COVID-19), represents a global problem for human health that involves the host lipid homeostasis. Regarding, lipid rafts are functional membrane microdomains with highly and tightly packed lipid molecules. These regions enriched in sphingolipids and cholesterol recruit and concentrate several receptors and molecules involved in pathogen recognition and cellular signaling. Cholesterol-rich lipid rafts have multiple functions for viral replication; however, their role in SARS-CoV-2 infection remains unclear. In this review, we discussed the novel evidence on the cholesterol-rich lipid rafts as a platform for SARS-CoV-2 entry, where receptors such as the angiotensin-converting enzyme-2 (ACE-2), heparan sulfate proteoglycans (HSPGs), human Toll-like receptors (TLRs), transmembrane serine proteases (TMPRSS), CD-147 and HDL-scavenger receptor B type 1 (SR-B1) are recruited for their interaction with the viral spike protein. FDA-approved drugs such as statins, metformin, hydroxychloroquine, and cyclodextrins (methyl-β-cyclodextrin) can disrupt cholesterol-rich lipid rafts to regulate key molecules in the immune signaling pathways triggered by SARS-CoV-2 infection. Taken together, better knowledge on cholesterol-rich lipid rafts in the SARS-CoV-2-host interactions will provide valuable insights into pathogenesis and the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Selvin Noé Palacios-Rápalo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Gustavo Martínez-Mier
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Judith Quistián-Galván
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Armando Muñoz-Pérez
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Víctor Bernal-Dolores
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Rosa María del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - José Manuel Reyes-Ruiz
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| |
Collapse
|
23
|
Farfan-Morales CN, Cordero-Rivera CD, Reyes-Ruiz JM, Hurtado-Monzón AM, Osuna-Ramos JF, González-González AM, De Jesús-González LA, Palacios-Rápalo SN, Del Ángel RM. Anti-flavivirus Properties of Lipid-Lowering Drugs. Front Physiol 2021; 12:749770. [PMID: 34690817 PMCID: PMC8529048 DOI: 10.3389/fphys.2021.749770] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Although Flaviviruses such as dengue (DENV) and zika (ZIKV) virus are important human pathogens, an effective vaccine or antiviral treatment against them is not available. Hence, the search for new strategies to control flavivirus infections is essential. Several studies have shown that the host lipid metabolism could be an antiviral target because cholesterol and other lipids are required during the replicative cycle of different Flaviviridae family members. FDA-approved drugs with hypolipidemic effects could be an alternative for treating flavivirus infections. However, a better understanding of the regulation between host lipid metabolism and signaling pathways triggered during these infections is required. The metabolic pathways related to lipid metabolism modified during DENV and ZIKV infection are analyzed in this review. Additionally, the role of lipid-lowering drugs as safe host-targeted antivirals is discussed.
Collapse
Affiliation(s)
- Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - José Manuel Reyes-Ruiz
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional "Adolfo Ruiz Cortines," Instituto Mexicano del Seguro Social, Heroica Veracruz, Mexico
| | - Arianna M Hurtado-Monzón
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Arely M González-González
- Laboratorio de Ingeniería Tisular y Medicina Traslacional, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Selvin Noé Palacios-Rápalo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Rosa María Del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
24
|
Madhivanan K, Ramadesikan S, Hsieh WC, Aguilar MC, Hanna CB, Bacallao RL, Aguilar RC. Lowe syndrome patient cells display mTOR- and RhoGTPase-dependent phenotypes alleviated by rapamycin and statins. Hum Mol Genet 2021; 29:1700-1715. [PMID: 32391547 DOI: 10.1093/hmg/ddaa086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/20/2020] [Accepted: 05/04/2020] [Indexed: 12/25/2022] Open
Abstract
Lowe syndrome (LS) is an X-linked developmental disease characterized by cognitive deficiencies, bilateral congenital cataracts and renal dysfunction. Unfortunately, this disease leads to the early death of affected children often due to kidney failure. Although this condition was first described in the early 1950s and the affected gene (OCRL1) was identified in the early 1990s, its pathophysiological mechanism is not fully understood and there is no LS-specific cure available to patients. Here we report two important signaling pathways affected in LS patient cells. While RhoGTPase signaling abnormalities led to adhesion and spreading defects as compared to normal controls, PI3K/mTOR hyperactivation interfered with primary cilia assembly (scenario also observed in other ciliopathies with compromised kidney function). Importantly, we identified two FDA-approved drugs able to ameliorate these phenotypes. Specifically, statins mitigated adhesion and spreading abnormalities while rapamycin facilitated ciliogenesis in LS patient cells. However, no single drug was able to alleviate both phenotypes. Based on these and other observations, we speculate that Ocrl1 has dual, independent functions supporting proper RhoGTPase and PI3K/mTOR signaling. Therefore, this study suggest that Ocrl1-deficiency leads to signaling defects likely to require combinatorial drug treatment to suppress patient phenotypes and symptoms.
Collapse
Affiliation(s)
- Kayalvizhi Madhivanan
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Swetha Ramadesikan
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Wen-Chieh Hsieh
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Mariana C Aguilar
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Claudia B Hanna
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| | - Robert L Bacallao
- Division of Nephrology, Indiana University School of Medicine, 340 W 10th St #6200, Indianapolis, IN 46202, USA
| | - R Claudio Aguilar
- Department of Biological Sciences, Purdue University, Hansen Life Sciences Building, Room 321, 201 S. University street, West Lafayette, IN 47907, USA
| |
Collapse
|
25
|
Inhibition of Orbivirus Replication by Fluvastatin and Identification of the Key Elements of the Mevalonate Pathway Involved. Viruses 2021; 13:v13081437. [PMID: 34452303 PMCID: PMC8402872 DOI: 10.3390/v13081437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 01/21/2023] Open
Abstract
Statin derivatives can inhibit the replication of a range of viruses, including hepatitis C virus (HCV, Hepacivirus), dengue virus (Flavivirus), African swine fever virus (Asfarviridae) and poliovirus (Picornaviridae). We assess the antiviral effect of fluvastatin in cells infected with orbiviruses (bluetongue virus (BTV) and Great Island virus (GIV)). The synthesis of orbivirus outer-capsid protein VP2 (detected by confocal immunofluorescence imaging) was used to assess levels of virus replication, showing a reduction in fluvastatin-treated cells. A reduction in virus titres of ~1.7 log (98%) in fluvastatin-treated cells was detected by a plaque assay. We have previously identified a fourth non-structural protein (NS4) of BTV and GIV, showing that it interacts with lipid droplets in infected cells. Fluvastatin, which inhibits 3-hydroxy 3-methyl glutaryl CoA reductase in the mevalonic acid pathway, disrupts these NS4 interactions. These findings highlight the role of the lipid pathways in orbivirus replication and suggest a greater role for the membrane-enveloped orbivirus particles than previously recognised. Chemical intermediates of the mevalonic acid pathway were used to assess their potential to rescue orbivirus replication. Pre-treatment of IFNAR(−/−) mice with fluvastatin promoted their survival upon challenge with live BTV, although only limited protection was observed.
Collapse
|
26
|
Development of Broad-Spectrum Antiviral Agents-Inspiration from Immunomodulatory Natural Products. Viruses 2021; 13:v13071257. [PMID: 34203182 PMCID: PMC8310077 DOI: 10.3390/v13071257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 01/04/2023] Open
Abstract
Developing broad-spectrum antiviral drugs remains an important issue as viral infections continue to threaten public health. Host-directed therapy is a method that focuses on potential targets in host cells or the body, instead of viral proteins. Its antiviral effects are achieved by disturbing the life cycles of pathogens or modulating immunity. In this review, we focus on the development of broad-spectrum antiviral drugs that enhance the immune response. Some natural products present antiviral effects mediated by enhancing immunity, and their structures and mechanisms are summarized here. Natural products with immunomodulatory effects are also discussed, although their antiviral effects remain unknown. Given the power of immunity and the feasibility of host-directed therapy, we argue that both of these categories of natural products provide clues that may be beneficial for the discovery of broad-spectrum antiviral drugs.
Collapse
|
27
|
Li H, Zhao Z, Li X, Qin L, Wen W, Chen H, Qian P. Cholesterol-25-Hydroxylase Suppresses Seneca Valley Virus Infection via Producing 25-Hydroxycholesterol to Block Adsorption Procedure. Virol Sin 2021; 36:1210-1219. [PMID: 34061318 DOI: 10.1007/s12250-021-00377-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/22/2020] [Indexed: 10/21/2022] Open
Abstract
Cholesterol-25-hydroxylase (CH25H) is a membrane protein associated with endoplasmic reticulum, and it is an interferon-stimulated factor regulated by interferon. CH25H catalyzes cholesterol to produce 25-hydroxycholesterol (25HC) by adding a second hydroxyl to the 25th carbon atom of cholesterol. Recent studies have shown that both CH25H and 25HC could inhibit the replication of many viruses. In this study, we found that ectopic expression of CH25H in HEK-293T and BHK-21 cell lines could inhibit the replication of Seneca Valley virus (SVV) and that there was no species difference. On the other hand, the knockdown of CH25H could enhance the replication of SVV in HEK-293T and BHK-21 cells, indicating the importance of CH25H. To some extent, the CH25H mutant without hydroxylase activity also lost its ability to inhibit SVV amplification. Further studies demonstrated that 25HC was involved in the entire life cycle of SVV, especially in repressing its adsorption process. This study reveals that CH25H exerts the advantage of innate immunity mainly by producing 25HC to block virion adsorption.
Collapse
Affiliation(s)
- Hui Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zekai Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiangmin Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Liuxing Qin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wei Wen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Ping Qian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China. .,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China. .,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
28
|
Proto MC, Fiore D, Piscopo C, Pagano C, Galgani M, Bruzzaniti S, Laezza C, Gazzerro P, Bifulco M. Lipid homeostasis and mevalonate pathway in COVID-19: Basic concepts and potential therapeutic targets. Prog Lipid Res 2021; 82:101099. [PMID: 33915202 PMCID: PMC8074527 DOI: 10.1016/j.plipres.2021.101099] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/21/2022]
Abstract
Despite encouraging progresses achieved in the management of viral diseases, efficient strategies to counteract infections are still required. The current global challenge highlighted the need to develop a rapid and cost-effective strategy to counteract the SARS-CoV-2 pandemic. Lipid metabolism plays a crucial role in viral infections. Viruses can use the host lipid machinery to support their life cycle and to impair the host immune response. The altered expression of mevalonate pathway-related genes, induced by several viruses, assures survival and spread in host tissue. In some infections, statins, HMG-CoA-reductase inhibitors, reduce cholesterol in the plasma membrane of permissive cells resulting in lower viral titers and failure to internalize the virus. Statins can also counteract viral infections through their immunomodulatory, anti-inflammatory and anti-thrombotic effects. Beyond statins, interfering with the mevalonate pathway could have an adjuvant effect in therapies aimed at mitigating endothelial dysfunction and deregulated inflammation in viral infection. In this review we depicted the historical and current evidence highlighting how lipid homeostasis and mevalonate pathway targeting represents a valid approach to rapidly neutralize viruses, focusing our attention to their potential use as effective targets to hinder SARS-CoV-2 morbidity and mortality. Pros and cons of statins and Mevalonate-pathway inhibitors have been also dissected.
Collapse
Affiliation(s)
- Maria Chiara Proto
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | - Chiara Piscopo
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy
| | - Cristina Pagano
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy
| | - Mario Galgani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy; Institute of Endocrinology and Experimental Oncology, IEOS CNR, 80131 Naples, Italy
| | - Sara Bruzzaniti
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, 80131 Naples, Italy; Department of Biology, University of Naples "Federico II", 80126 Naples, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, 80131 Naples, Italy
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy.
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| |
Collapse
|
29
|
Zhu L, Liu F, Hao Q, Feng T, Chen Z, Luo S, Xiao R, Sun M, Zhang T, Fan X, Zeng X, He J, Yuan P, Liu J, Ruiz M, Dupuis J, Hu Q. Dietary Geranylgeranyl Pyrophosphate Counteracts the Benefits of Statin Therapy in Experimental Pulmonary Hypertension. Circulation 2021; 143:1775-1792. [PMID: 33660517 DOI: 10.1161/circulationaha.120.046542] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND The mevalonate pathway generates endogenous cholesterol and intermediates including geranylgeranyl pyrophosphate (GGPP). By reducing GGPP production, statins exert pleiotropic or cholesterol-independent effects. The potential regulation of GGPP homeostasis through dietary intake and the interaction with concomitant statin therapy is unknown. METHODS We developed a sensitive high-pressure liquid chromatography technique to quantify dietary GGPP and conducted proteomics, qualitative real-time polymerase chain reaction screening, and Western blot to determine signaling cascades, gene expression, protein-protein interaction, and protein membrane trafficking in wild-type and transgenic rats. RESULTS GGPP contents were highly variable depending on food source that differentially regulated blood GGPP levels in rats. Diets containing intermediate and high GGPP reduced or abolished the effects of statins in rats with hypoxia- and monocrotaline-induced pulmonary hypertension: this was rescuable by methyl-allylthiosulfinate and methyl-allylthiosulfinate-rich garlic extracts. In human pulmonary artery smooth muscle cells treated with statins, hypoxia activated RhoA in an extracellular GGPP-dependent manner. Hypoxia-induced ROCK2 (Rho associated coiled-coil containing protein kinase 2)/Rab10 (Ras-related protein rab-10) signaling was prevented by statin and recovered by exogenous GGPP. The hypoxia-activated RhoA/ROCK2 pathway in rat and human pulmonary artery smooth muscle cells upregulated the expression of Ca2+-sensing receptor (CaSR) and HIMF (hypoxia-induced mitogenic factor), a mechanism attenuated by statin treatment and regained with exogenous GGPP. Rab10 knockdown almost abrogated hypoxia-promoted CaSR membrane trafficking, a process diminished by statin and resumed by exogenous GGPP. Hypoxia-induced pulmonary hypertension was reduced in rats with CaSR mutated at the binding motif of HIMF and the interaction between dietary GGPP and statin efficiency was abolished. In humans fed a high GGPP diet, blood GGPP levels were increased. This abolished statin-lowering effects on plasma GGPP, and also on hypoxia-enhanced RhoA activity of blood monocytes that was rescued by garlic extracts. CONCLUSIONS There is important dietary regulation of GGPP levels that interferes with the effects of statin therapy in experimental pulmonary hypertension. These observations rely on a key and central role of RhoA-ROCK2 cascade activation and Rab10-faciliated CaSR membrane trafficking with subsequent overexpression and binding of HIMF to CaSR. These findings warrant clinical investigation for the treatment of pulmonary hypertension and perhaps other diseases by combining statin with garlic-derived methyl-allylthiosulfinate or garlic extracts and thus circumventing dietary GGPP variations.
Collapse
Affiliation(s)
- Liping Zhu
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangbo Liu
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Hao
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian Feng
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeshuai Chen
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengquan Luo
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xiao
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengxiang Sun
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Zhang
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohang Fan
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianqin Zeng
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianguo He
- State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.H.)
| | - Ping Yuan
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, China (P.Y., J.L.)
| | - Jinming Liu
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, China (P.Y., J.L.)
| | - Matthieu Ruiz
- Departments of Nutrition (M.R.), Université de Montréal, Canada.,Montreal Heart Institute Research Center, Canada (M.R., J.D.)
| | - Jocelyn Dupuis
- Medicine (J.D.), Université de Montréal, Canada.,Montreal Heart Institute Research Center, Canada (M.R., J.D.)
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Ministry of Health (L.Z., F.L., Q. Hao, T.F., Z.C., S.L., R.X., M.S., T.Z., X.F., X.Z., Q. Hu), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Fares A, Borrmann D, Ivester JR. Are statins beneficial for the treatment of SARS-CoV-2 infection? J Infect Prev 2021; 22:177-180. [PMID: 34295380 DOI: 10.1177/1757177420982049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/08/2020] [Indexed: 12/20/2022] Open
Abstract
Novel coronavirus disease 2019 (COVID-19) is a highly infectious, rapidly spreading viral disease and has emerged as a public health emergency of international concern. As of this time, there are no specific antiviral therapies available for the treatment of COVID-19. However, it is possible that some existing drugs, usually used for other conditions, may have some benefits. Statins have been widely reported to exert antiviral activity against many enveloped viruses by inhibiting the cholesterol biosynthesis pathway. Cholesterol likewise contributes to the coronavirus's life cycle, including viral entry, fusion and budding. In addition, statins have been ascribed beneficial anti-inflammatory, immunomodulatory effects and promote haemodynamic stability. Therefore, statins, which are cholesterol-lowering drugs with anti-inflammatory, immunomodulatory and antiviral properties, may play a role in SARS-CoV-2 therapy. The aim of the present minireview was to delineate the potential beneficial therapeutic effects of statins in treating SARS-CoV-2 infections. Nevertheless, large, randomised trials are needed to confirm the beneficial effects and safety profile of the statins in patients with SARS-CoV-2.
Collapse
Affiliation(s)
- Auda Fares
- St. Willibrord- Spital Emmerich-Rees Hospital, Acute Geriatrics Medicine and Rehabilitation, Emmerich am Rhein/ Germany
| | - Dieter Borrmann
- St. Willibrord- Spital Emmerich-Rees Hospital, Acute Geriatrics Medicine and Rehabilitation, Emmerich am Rhein/ Germany
| | - Julius R Ivester
- Department of Anaesthesia and Pain Management, Roper Hospital, Charleston, SC, USA
| |
Collapse
|
31
|
|
32
|
Ogrinc K, Kastrin A, Lotrič-Furlan S, Bogovič P, Rojko T, Cerar-Kišek T, Ružić-Sabljić E, Wormser GP, Strle F. Effect of Statin Use on the Clinical Manifestations, Laboratory Test Results and Outcome of Lyme Neuroborreliosis. J Clin Med 2020; 9:jcm9092995. [PMID: 32948002 PMCID: PMC7563736 DOI: 10.3390/jcm9092995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
Statins have anti-inflammatory and potentially antimicrobial activity, but whether they have a beneficial effect on the course of infectious diseases is controversial. In this study, we assessed the impact of pre-existing statin use on the course and outcome of Lyme neuroborreliosis manifested as meningoradiculitis (Bannwarth’s syndrome). One hundred and twenty three consecutive patients with Bannwarth’s syndrome, of whom 18 (14.6%) were being treated with statins, were included in the study. To assess the influence of statin use on the course and outcome of the disease, univariate and multivariable analyses were performed. No statistically significant association was found between statin pre-treatment and the clinical manifestations, laboratory test results, and outcome of Bannwarth’s syndrome. In conclusion, pre-existing use of statins did not significantly impact either the clinical presentation or the outcome of Bannwarth’s syndrome.
Collapse
Affiliation(s)
- Katarina Ogrinc
- Department of Infectious Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia; (S.L.-F.); (P.B.); (T.R.); (F.S.)
- Correspondence: ; Tel.: +386-1522-2110
| | - Andrej Kastrin
- Institute for Biostatistics and Medical Informatics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Stanka Lotrič-Furlan
- Department of Infectious Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia; (S.L.-F.); (P.B.); (T.R.); (F.S.)
| | - Petra Bogovič
- Department of Infectious Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia; (S.L.-F.); (P.B.); (T.R.); (F.S.)
| | - Tereza Rojko
- Department of Infectious Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia; (S.L.-F.); (P.B.); (T.R.); (F.S.)
| | - Tjaša Cerar-Kišek
- Institute for Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.C.-K.); (E.R.-S.)
| | - Eva Ružić-Sabljić
- Institute for Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.C.-K.); (E.R.-S.)
| | - Gary P. Wormser
- Division of Infectious Diseases, New York Medical College, Valhalla, NY 10595, USA;
| | - Franc Strle
- Department of Infectious Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia; (S.L.-F.); (P.B.); (T.R.); (F.S.)
| |
Collapse
|
33
|
Gorabi AM, Kiaie N, Bianconi V, Jamialahmadi T, Al-Rasadi K, Johnston TP, Pirro M, Sahebkar A. Antiviral effects of statins. Prog Lipid Res 2020; 79:101054. [PMID: 32777243 DOI: 10.1016/j.plipres.2020.101054] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Introducing statins as possible widely-available drugs for the treatment of viral infections requires an in depth review of their antiviral properties. Despite some inconsistency, a large body of literature data from experimental and clinical studies suggest that statins may have a role in the treatment of viral infections due to their immunomodulatory properties as well as their ability to inhibit viral replication. In the present review, the role that statins may play while interacting with the immune system during viral infections and the possible inhibitory effects of statins on different stages of virus cell cycle (i.e., from fusion with host cell membranes to extracellular release) and subsequent virus transmission are described. Specifically, cholesterol-dependent and cholesterol-independent mechanisms of the antiviral effects of statins are reported.
Collapse
Affiliation(s)
- Armita M Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Kiaie
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Tannaz Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalid Al-Rasadi
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy.
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| |
Collapse
|
34
|
Li S, Li L, Zhu H, Shi M, Fan H, Gao Y, Wang X, Jiang P, Bai J. Cholesterol 25-hydroxylase inhibits encephalomyocarditis virus replication through enzyme activity-dependent and independent mechanisms. Vet Microbiol 2020; 245:108658. [PMID: 32456829 DOI: 10.1016/j.vetmic.2020.108658] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/19/2020] [Accepted: 03/22/2020] [Indexed: 12/19/2022]
Abstract
Cholesterol-25-hydroxylase (CH25 H) is a reticulum-associated membrane protein induced by an important interferon-stimulating gene (ISG) and can significantly inhibit some virus replication. But the effect of CH25H on encephalomyocarditis virus (EMCV) is still not clear. In this study, we found that EMCV infection increases significantly the endogenous CH25H expression in BHK-21 and N2a cells. CH25H and cholesterol catalytic oxidation product 25-hydroxycholesterol (25HC) obviously inhibits EMCV infection by inhibiting the viral penetration. But the CH25H mutant lacking hydroxylase activity repairs the ability to inhibit the viral replication. Meanwhile, β-cyclodextrin crystalline as a cholesterol inhibitor significantly decreases the viral replication. In addition, CH25H can selectively interact and degrade the viral RNA-Dependent RNA Polymerase-3D protein by independent on the association of proteasome, lysosome and caspase manner. It provides new insights into the interplay mechanisms between CH25H and non-enveloped single-stranded positive RNA viruses.
Collapse
Affiliation(s)
- Shihai Li
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Liang Li
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Huixin Zhu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Mengyu Shi
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Hui Fan
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - YanNi Gao
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - XianWei Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, PR China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, PR China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
35
|
Rosuvastatin Enhances VSV-G Lentiviral Transduction of NK Cells via Upregulation of the Low-Density Lipoprotein Receptor. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:634-646. [PMID: 32300610 PMCID: PMC7150439 DOI: 10.1016/j.omtm.2020.03.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/23/2020] [Indexed: 01/20/2023]
Abstract
Adoptive natural killer (NK) cell therapy is attaining promising clinical outcomes in recent years, but improvements are needed. Genetic modification of NK cells with a tumor antigen-specific receptor on their surface coupled to intracellular signaling domains may lead to enhanced cytotoxicity against malignant cells. One of the most common approaches is by lentivirus-mediated transduction. However, NK cells are difficult to transduce and various methods have been attempted with different success rates. Because the low-density lipoprotein-receptor (LDLR) is the receptor of vesicular stomatitis virus (VSV) and is expressed only at low levels on NK cells, we tested the potential of 5 statins and 5 non-statin compounds to increase the LDLR expression, thereby facilitating viral transduction. We found that the transduction efficiency of VSV-G pseudotyped lentivirus is augmented by statins that induced higher LDLR expression. In both NK-92 cells and primary NK cells, the transduction efficiency increased after treatment with statins. Furthermore, statins have been reported to suppress NK cell cytotoxicity; however, we showed that this can be completely reversed by adding geranylgeranyl-pyrophosphate (GGPP). Among the statins tested, we found that the combination of rosuvastatin with GGPP most potently improved viral transduction without affecting the cytotoxic properties of the NK cells.
Collapse
|
36
|
Abstract
Apolipoprotein A-I binding protein (AIBP) is a recently identified innate anti-inflammatory factor. Here, we show that AIBP inhibited HIV replication by targeting lipid rafts and reducing virus-cell fusion. Importantly, AIBP selectively reduced levels of rafts on cells stimulated by an inflammatory stimulus or treated with extracellular vesicles containing HIV-1 protein Nef without affecting rafts on nonactivated cells. Accordingly, fusion of monocyte-derived macrophages with HIV was sensitive to AIBP only in the presence of Nef. Silencing of endogenous AIBP significantly upregulated HIV-1 replication. Interestingly, HIV-1 replication in cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, was not inhibited by AIBP. These results suggest that AIBP is an innate anti-HIV factor that targets virus-cell fusion. Apolipoprotein A-I binding protein (AIBP) is a protein involved in regulation of lipid rafts and cholesterol efflux. AIBP has been suggested to function as a protective factor under several sets of pathological conditions associated with increased abundance of lipid rafts, such as atherosclerosis and acute lung injury. Here, we show that exogenously added AIBP reduced the abundance of lipid rafts and inhibited HIV replication in vitro as well as in HIV-infected humanized mice, whereas knockdown of endogenous AIBP increased HIV replication. Endogenous AIBP was much more abundant in activated T cells than in monocyte-derived macrophages (MDMs), and exogenous AIBP was much less effective in T cells than in MDMs. AIBP inhibited virus-cell fusion, specifically targeting cells with lipid rafts mobilized by cell activation or Nef-containing exosomes. MDM-HIV fusion was sensitive to AIBP only in the presence of Nef provided by the virus or exosomes. Peripheral blood mononuclear cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, bound less AIBP than cells from donors with other HLA genotypes and were not protected by AIBP from rapid HIV-1 replication. These results provide the first evidence for the role of Nef exosomes in regulating HIV-cell fusion by modifying lipid rafts and suggest that AIBP is an innate factor that restricts HIV replication by targeting lipid rafts.
Collapse
|
37
|
Parihar SP, Guler R, Brombacher F. Statins: a viable candidate for host-directed therapy against infectious diseases. Nat Rev Immunol 2019; 19:104-117. [PMID: 30487528 DOI: 10.1038/s41577-018-0094-3] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Statins were first identified over 40 years ago as lipid-lowering drugs and have been remarkably effective in treating cardiovascular diseases. As research advanced, the protective effects of statins were additionally attributed to their anti-inflammatory, antioxidative, anti-thrombotic and immunomodulatory functions rather than lipid-lowering abilities alone. By promoting host defence mechanisms and inhibiting pathological inflammation, statins increase survival in human infectious diseases. At the cellular level, statins inhibit the intermediates of the host mevalonate pathway, thus compromising the immune evasion strategies of pathogens and their survival. Here, we discuss the potential use of statins as an inexpensive and practical alternative or adjunctive host-directed therapy for infectious diseases caused by intracellular pathogens, such as viruses, protozoa, fungi and bacteria.
Collapse
Affiliation(s)
- Suraj P Parihar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa. .,Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC), Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Division of Medical Microbiology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
| | - Reto Guler
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa.,Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC), Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, Cape Town, South Africa. .,Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC), Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
38
|
Fatty Acids Regulate Porcine Reproductive and Respiratory Syndrome Virus Infection via the AMPK-ACC1 Signaling Pathway. Viruses 2019; 11:v11121145. [PMID: 31835577 PMCID: PMC6950460 DOI: 10.3390/v11121145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 12/24/2022] Open
Abstract
Lipids play a crucial role in the replication of porcine reproductive and respiratory syndrome virus (PRRSV), a porcine virus that is endemic throughout the world. However, little is known about the effect of fatty acids (FAs), a type of vital lipid, on PRRSV infection. In this study, we found that treatment with a FA biosynthetic inhibitor significantly inhibited PRRSV propagation, indicating the necessity of FAs for optimal replication of PRRSV. Further study revealed that 5′-adenosine monophosphate (AMP)-activated protein kinase (AMPK), a key kinase antagonizing FA biosynthesis, was strongly activated by PRRSV and the pharmacological activator of AMPK exhibited anti-PRRSV activity. Additionally, we found that acetyl-CoA carboxylase 1 (ACC1), the first rate-limiting enzyme in the FA biosynthesis pathway, was phosphorylated (inactive form) by PRRSV-activated AMPK, and active ACC1 was required for PRRSV proliferation, suggesting that the PRRSV infection induced the activation of the AMPK–ACC1 pathway, which was not conducive to PRRSV replication. This work provides new evidence about the mechanisms involved in host lipid metabolism during PRRSV infection and identifies novel potential antiviral targets for PRRSV.
Collapse
|
39
|
Du X, Zuo X, Meng F, Wu F, Zhao X, Li C, Cheng G, Qin FXF. Combinatorial screening of a panel of FDA-approved drugs identifies several candidates with anti-Ebola activities. Biochem Biophys Res Commun 2019; 522:862-868. [PMID: 31806372 DOI: 10.1016/j.bbrc.2019.11.065] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/10/2019] [Indexed: 01/16/2023]
Abstract
Ebola virus (EBOV), pathogen of Ebola hemorrhagic fever (EHF), is an enveloped filamental RNA virus. Recently, the EHF crisis occurred in the Democratic Republic of the Congo again highlights the urgency for its clinical treatments. However, no Food and Drug Administration (FDA)-approved therapeutics are currently available. Drug repurposing screening is a time- and cost-effective approach for identifying anti-EBOV therapeutics. Here, by combinatorial screening using pseudovirion and minigenome replicon systems we have identified several FDA-approved drugs with significant anti-EBOV activities. These potential candidates include azithromycin, clomiphene, chloroquine, digitoxin, epigallocatechin-gallate, fluvastatin, tetrandrine and tamoxifen. Mechanistic studies revealed that fluvastatin inhibited EBOV pseudovirion entry by blocking the pathway of mevalonate biosynthesis, while the inhibitory effect of azithromycin on EBOV maybe due to its intrinsic cationic amphiphilic structure altering the homeostasis of later endosomal vesicle similar as tamoxifen. Moreover, based on structure and pathway analyses, the anti-EBOV activity has been extended to other family members of statins, such as simvastatin, and multiple other cardiac glycoside drugs, some of which exhibited even stronger activities. More importantly, in searching for drug interaction, we found various synergy between several anti-EBOV drug combinations, showing substantial and powerful synergistic against EBOV infection. In conclusion, our work illustrates a successful and productive approach to identify new mechanisms and targets for treating EBOV infection by combinatorial screening of FDA-approved drugs.
Collapse
Affiliation(s)
- Xiaohong Du
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China
| | - Xiangyang Zuo
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China
| | - Fang Meng
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China
| | - Fei Wu
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China
| | - Xin Zhao
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China
| | - Chunfeng Li
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China
| | - Genhong Cheng
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - F Xiao-Feng Qin
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China; Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
40
|
Liu B, Zhang X, Zhang W, Wu L, Jing S, Liu W, Xia B, Zou F, Lu L, Ma X, He D, Hu Q, Zhang Y, Deng K, Cai W, Tang X, Peng T, Zhang H, Li L. Lovastatin Inhibits HIV-1-Induced MHC-I Downregulation by Targeting Nef-AP-1 Complex Formation: A New Strategy to Boost Immune Eradication of HIV-1 Infected Cells. Front Immunol 2019; 10:2151. [PMID: 31572371 PMCID: PMC6749138 DOI: 10.3389/fimmu.2019.02151] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/27/2019] [Indexed: 01/05/2023] Open
Abstract
Current combined antiretroviral therapy (cART) mainly targets 3 of the 15 HIV proteins leaving many potential viral vulnerabilities unexploited. To purge the HIV-1 latent reservoir, various strategies including “shock and kill” have been developed. A key question is how to restore impaired immune surveillance. HIV-1 protein Nef has long been known to mediate the downregulation of cell-surface MHC-I and assist HIV-1 to evade the immune system. Through high throughput screening of Food and Drug Administration (FDA) approved drugs, we identified lovastatin, a statin drug, to significantly antagonize Nef to downregulate MHC-I, CD4, and SERINC5, and inhibit the intrinsic infectivity of virions. In addition, lovastatin boosted autologous CTLs to eradicate the infected cells and effectively inhibit the subsequent viral rebound in CD4+ T-lymphocytes isolated from HIV-1-infected individuals receiving suppressive cART. Furthermore, we found that lovastatin inhibits Nef-induced MHC-I downregulation by directly binding with Nef and disrupting the Nef–AP-1 complex. These results demonstrate that lovastatin is a promising agent for counteracting Nef-mediated downregulation of MHC-I, CD4, and SERINC5. Lovastatin could potentially be used in the clinic to enhance anti-HIV-1 immune surveillance.
Collapse
Affiliation(s)
- Bingfeng Liu
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China.,Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xu Zhang
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Wanying Zhang
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Liyang Wu
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Shuliang Jing
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Liu
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Baijin Xia
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Fan Zou
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China.,Department of Molecular Therapy, Qianyang Biomedical Research Institute, Guangzhou, China.,Guangzhou Women and Children Hospital, Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Lijuan Lu
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Xiancai Ma
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Dalian He
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Qifei Hu
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China.,Department of Molecular Therapy, Qianyang Biomedical Research Institute, Guangzhou, China
| | - Yiwen Zhang
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Kai Deng
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Weiping Cai
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoping Tang
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tao Peng
- Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Hui Zhang
- Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Institute of Human Virology, Sun Yat-sen University, Guangzhou, China.,Department of Molecular Therapy, Qianyang Biomedical Research Institute, Guangzhou, China
| | - Linghua Li
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
41
|
Yu X, Shang H, Jiang Y. ICAM-1 in HIV infection and underlying mechanisms. Cytokine 2019; 125:154830. [PMID: 31491723 DOI: 10.1016/j.cyto.2019.154830] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/29/2019] [Accepted: 08/24/2019] [Indexed: 02/07/2023]
Abstract
Intercellular adhesion molecule 1 (ICAM-1) is a glycoprotein that participates in inflammatory and immune responses. Both cell surface and soluble ICAM-1 are significantly increased during human immunodeficiency virus (HIV) infection, and ICAM-1 has important functions in promoting inflammatory responses and enhancing HIV infectivity; however, a comprehensive summary these roles has yet to be elaborated. In this review we describe the general biological characteristics of ICAM-1, its association with HIV disease progression and promotion of HIV production, mechanisms inducing upregulation of ICAM-1, and possible intervention strategies, representing important insights in the context of HIV treatment.
Collapse
Affiliation(s)
- Xiaowen Yu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China.
| | - Yongjun Jiang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang 110001, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou 310003, China.
| |
Collapse
|
42
|
Españo E, Nam JH, Song EJ, Song D, Lee CK, Kim JK. Lipophilic statins inhibit Zika virus production in Vero cells. Sci Rep 2019; 9:11461. [PMID: 31391514 PMCID: PMC6685969 DOI: 10.1038/s41598-019-47956-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/25/2019] [Indexed: 11/09/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne member of the Flaviviridae family. ZIKV infection has been associated with neurological complications such as microcephaly in newborns and Guillain-Barré syndrome in adults; thus, therapeutic agents are urgently needed. Statins are clinically approved for lowering cholesterol levels to prevent cardiovascular disease but have shown potential as antiviral drugs. In this study, we explored the possibility of utilizing statins as anti-ZIKV drugs. We found that, generally, lipophilic statins (atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, and simvastatin) could reduce ZIKV production in vitro and result in smaller foci of infection. Time-of-drug-addition assay revealed that early treatment with statins is more beneficial than late treatment; however, statins could not completely inhibit the entry stage of ZIKV infection. Furthermore, individual lipophilic statins differed in anti-ZIKV capacity, with fluvastatin being the most efficient at low concentrations. Taken together, this study shows that statins or their derivatives have the potential to be used as anti-ZIKV therapeutic agents.
Collapse
Affiliation(s)
- Erica Españo
- Department of Pharmacy, Korea University College of Pharmacy, Sejong, 30019, Republic of Korea
| | - Jeong-Hyun Nam
- Department of Pharmacy, Korea University College of Pharmacy, Sejong, 30019, Republic of Korea
| | - Eun-Jung Song
- Department of Pharmacy, Korea University College of Pharmacy, Sejong, 30019, Republic of Korea
| | - Daesub Song
- Department of Pharmacy, Korea University College of Pharmacy, Sejong, 30019, Republic of Korea
| | - Chong-Kil Lee
- Department of Pharmacy, College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, 28160, Republic of Korea.
| | - Jeong-Ki Kim
- Department of Pharmacy, Korea University College of Pharmacy, Sejong, 30019, Republic of Korea.
| |
Collapse
|
43
|
McCrae C, Dzgoev A, Ståhlman M, Horndahl J, Svärd R, Große A, Großkopf T, Skujat MA, Williams N, Schubert S, Echeverri C, Jackson C, Guedán A, Solari R, Vaarala O, Kraan M, Rådinger M. Lanosterol Synthase Regulates Human Rhinovirus Replication in Human Bronchial Epithelial Cells. Am J Respir Cell Mol Biol 2019; 59:713-722. [PMID: 30084659 DOI: 10.1165/rcmb.2017-0438oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human rhinovirus (RV) infections are a significant risk factor for exacerbations of asthma and chronic obstructive pulmonary disease. Thus, approaches to prevent RV infection in such patients would give significant benefit. Through RNA interference library screening, we identified lanosterol synthase (LSS), a component of the cholesterol biosynthetic pathway, as a novel regulator of RV replication in primary normal human bronchial epithelial cells. Selective knock down of LSS mRNA with short interfering RNA inhibited RV2 replication in normal human bronchial epithelial cells. Small molecule inhibitors of LSS mimicked the effect of LSS mRNA knockdown in a concentration-dependent manner. We further demonstrated that the antiviral effect is not dependent on a reduction in total cellular cholesterol but requires a 24-hour preincubation with the LSS inhibitor. The rank order of antiviral potency of the LSS inhibitors used was consistent with LSS inhibition potency; however, all compounds showed remarkably higher potency against RV compared with the LSS enzyme potency. We showed that LSS inhibition led to an induction of 24(S),25 epoxycholesterol, an important regulator of the sterol pathway. We also demonstrated that LSS inhibition led to a profound increase in expression of the innate antiviral defense protein, IFN-β. We found LSS to be a novel regulator of RV replication and innate antiviral immunity and identified a potential molecular mechanism for this effect, via induction of 24(S),25 epoxycholesterol. Inhibition of LSS could therefore be a novel therapeutic target for prevention of RV-induced exacerbations.
Collapse
Affiliation(s)
- Christopher McCrae
- 1 Respiratory Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden.,2 Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, and
| | - Anatoly Dzgoev
- 1 Respiratory Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Marcus Ståhlman
- 3 Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Horndahl
- 1 Respiratory Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Rebecka Svärd
- 1 Respiratory Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | | | | | | - Nicola Williams
- 5 AstraZeneca Research and Development, Charnwood, Loughborough, United Kingdom
| | | | | | - Clive Jackson
- 5 AstraZeneca Research and Development, Charnwood, Loughborough, United Kingdom
| | - Anabel Guedán
- 7 Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Roberto Solari
- 7 Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Outi Vaarala
- 1 Respiratory Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Maarten Kraan
- 1 Respiratory Inflammation and Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Madeleine Rådinger
- 2 Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, and
| |
Collapse
|
44
|
Relaño-Rodríguez I, Juárez-Sánchez R, Pavicic C, Muñoz E, Muñoz-Fernández MÁ. Polyanionic carbosilane dendrimers as a new adjuvant in combination with latency reversal agents for HIV treatment. J Nanobiotechnology 2019; 17:69. [PMID: 31113488 PMCID: PMC6529996 DOI: 10.1186/s12951-019-0500-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
Background The major obstacle impeding human immunodeficiency virus-1 (HIV-1) eradication in antiretroviral treatment (ART) treated HIV-1 subjects is the establishment of long-lived latently infected resting CD4+ T cells. Due to the fact that no drug has been effective, the search for new drugs and combinations are a priority in the HIV cure. Treatments based on nanotechnology have emerged as an innovative and promising alternative to current and conventional therapies. In this respect, nanotechnology opens up a new door for eliminating latent HIV infection. We studied the role of G1-S4, G2-S16 and G3-S16 polyanionic carbosilane dendrimers in the context of latent HIV-1 persistence. Moreover, we study the efficiency of these dendrimers in combination with latency reversal agents (LRAs) against HIV-1 infection. Methods J89GFP lymphocyte and THP89GFP monocyte derived cell lines latently infected with HIV-1 p89GFP were used as an in vitro model of latency for our study. Viability assays by 3-(4-5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) were performed to determine the working concentrations of dendrimers and LRAs. Both cell lines were treated with G1-S4, G2-S16 and G3-S16 either alone or in combination with bryostatin (BRY), romidepsin (RMD) or panobinostat (PNB) for 24 and 48 h. The expression pattern of GFP was measured by flow cytometry and referred as measure of viral reactivation. Results and discussion The combination treatment of the dendrimers with the protein kinase C (PKC) agonist did not modify the antilatency activity in J89GFP lymphocyte cell line. Interestingly enough, G3-S16 dendrimer alone and its combination with BRY, RMD or PNB showed a significant increased expression of GFP in the THP89GFP monocyte cell line. Conclusion We showed for the first time that nanoparticles, in this case, G3-S16 anionic carbosilan dendrimer may play an important role in new treatments against HIV-1 infection.![]()
Collapse
Affiliation(s)
- Ignacio Relaño-Rodríguez
- Molecular Immunology Laboratory, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Health Research Institute Gregorio Marañón (IiSGM), Spanish HIV HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Innohealth, Parque Científico de Madrid, Madrid, Spain
| | - Raquel Juárez-Sánchez
- Molecular Immunology Laboratory, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Health Research Institute Gregorio Marañón (IiSGM), Spanish HIV HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | | | - Eduardo Muñoz
- Department of Cell Biology, Physiology and Immunology, Instituto Maimónides de Investigaciones Biomédicas de Córdoba (IMIBIC)/Reina Sofia University Hospital University of Córdoba, Córdoba, Spain
| | - Maria Ángeles Muñoz-Fernández
- Molecular Immunology Laboratory, Hospital General Universitario Gregorio Marañón, Madrid, Spain. .,Health Research Institute Gregorio Marañón (IiSGM), Spanish HIV HGM BioBank, Madrid, Spain. .,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
45
|
Mesquita I, Estaquier J. Viral Manipulation of the Host Metabolic Network. EXPERIENTIA. SUPPLEMENTUM 2019; 109:377-401. [PMID: 30535606 DOI: 10.1007/978-3-319-74932-7_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Viruses are intracellular parasites that rely on host machinery to replicate and achieve a successful infection. Viruses have evolved to retain a broad range of strategies to manipulate host cell metabolism and metabolic resources, channeling them toward the production of virion components leading to viral production. Although several viruses share similar strategies for manipulating host cell metabolism, these processes depend on several factors, namely, the viral life cycle and the metabolic and energetic status of the infected cell. Based on this knowledge, the development of new therapeutic approaches that circumvent viral spread through the target of altered metabolic pathways is an opportunity to tackle the infection. However, finding effective broad-spectrum strategies that aim at restoring to homeostasis the metabolic alterations induced upon virus infection is still a Holy Grail quest for antiviral therapies. Here, we review the strategies by which viruses manipulate host metabolism for their own benefit, with a particular emphasis on carbohydrate, glutamine, and lipid metabolism.
Collapse
Affiliation(s)
- Inês Mesquita
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Jérôme Estaquier
- Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada. .,CNRS FR 3636, Université Paris Descartes, Paris, France.
| |
Collapse
|
46
|
Host-directed kinase inhibitors act as novel therapies against intracellular Staphylococcus aureus. Sci Rep 2019; 9:4876. [PMID: 30890742 PMCID: PMC6425000 DOI: 10.1038/s41598-019-41260-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/05/2019] [Indexed: 01/09/2023] Open
Abstract
Host-directed therapeutics are a promising anti-infective strategy against intracellular bacterial pathogens. Repurposing host-targeted drugs approved by the FDA in the US, the MHRA in the UK and/or regulatory equivalents in other countries, is particularly interesting because these drugs are commercially available, safe doses are documented and they have been already approved for other clinical purposes. In this study, we aimed to identify novel therapies against intracellular Staphylococcus aureus, an opportunistic pathogen that is able to exploit host molecular and metabolic pathways to support its own intracellular survival. We screened 133 host-targeting drugs and found three host-directed tyrosine kinase inhibitors (Ibrutinib, Dasatinib and Crizotinib) that substantially impaired intracellular bacterial survival. We found that Ibrutinib significantly increased host cell viability after S. aureus infection via inhibition of cell invasion and intracellular bacterial proliferation. Using phosphoproteomics data, we propose a putative mechanism of action of Ibrutinib involving several host factors, including EPHA2, C-JUN and NWASP. We confirmed the importance of EPHA2 for staphylococcal infection in an EPHA2-knock-out cell line. Our study serves as an important example of feasibility for identifying host-directed therapeutics as candidates for repurposing.
Collapse
|
47
|
Guerra-De-Blas PDC, Torres-González P, Bobadilla-Del-Valle M, Sada-Ovalle I, Ponce-De-León-Garduño A, Sifuentes-Osornio J. Potential Effect of Statins on Mycobacterium tuberculosis Infection. J Immunol Res 2018; 2018:7617023. [PMID: 30581876 PMCID: PMC6276473 DOI: 10.1155/2018/7617023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/11/2018] [Accepted: 10/23/2018] [Indexed: 02/03/2023] Open
Abstract
Tuberculosis is one of the 10 leading causes of death in the world. The current treatment is based on a combination of antimicrobials administered for six months. It is essential to find therapeutic agents with which the treatment time can be shortened and strengthen the host immune response against Mycobacterium tuberculosis. M. tuberculosis needs cholesterol to infect and survive inside the host, but the progression of the infection depends to a large extent on the capacity of the immune response to contain the infection. Statins inhibit the synthesis of cholesterol and have pleiotropic effects on the immune system, which have been associated with better results in the treatment of several infectious diseases. Recently, it has been reported that cells treated with statins are more resistant to M. tuberculosis infection, and they have even been proposed as adjuvants in the treatment of M. tuberculosis infection. The aim of this review is to summarize the immunopathogenesis of tuberculosis and its mechanisms of evasion and to compile the available scientific information on the effect of statins in the treatment of tuberculosis.
Collapse
Affiliation(s)
- Paola Del Carmen Guerra-De-Blas
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Pedro Torres-González
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miriam Bobadilla-Del-Valle
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Isabel Sada-Ovalle
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Alfredo Ponce-De-León-Garduño
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Sifuentes-Osornio
- Laboratory of Clinical Microbiology, Department of Infectious Diseases, Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
48
|
Impact of pre-existing treatment with statins on the course and outcome of tick-borne encephalitis. PLoS One 2018; 13:e0204773. [PMID: 30286159 PMCID: PMC6171849 DOI: 10.1371/journal.pone.0204773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Although statins have anti-inflammatory and potentially also antimicrobial (including antiviral) activity, their therapeutic impact on infectious diseases is controversial. In this study, we evaluated whether pre-existing statin use influenced the course and outcome of tick-borne encephalitis. METHODS To assess the influence of statin usage on the severity of acute illness and the outcome of tick-borne encephalitis, univariate and multivariable analyses were performed for 700 adult patients with tick-borne encephalitis of whom 77 (11%) were being treated with statins, and for 410 patients of whom 53 (13%) were receiving statins, respectively. RESULTS Multivariable analyses found no statistically significant association between statin usage and having a milder acute illness. There was also no statistically significant benefit with respect to a favorable outcome defined by the absence of post-encephalitic syndrome (ORs for a favorable outcome at 6 months was 0.96, 95% CI: 0.46-2.04, P = 0.926; at 12 months 0.29, 95% CI: 0.06-1.33, P = 0.111; at 2-7 years after acute illness 0.44, 95% CI: 0.09-2.22, P = 0.321), by a reduction in the frequency of six nonspecific symptoms (fatigue, myalgia/arthralgia memory disturbances, headache, concentration disturbances, irritability) occurring during the 4 week period before the last examination, or by higher SF-36 scores in any of the eight separate domains of health as well as in the physical and mental global overall component. Furthermore, there were no significant differences between patients receiving statins and those who were not in the cerebrospinal fluid or serum levels for any of the 24 cytokines/chemokines measured. CONCLUSIONS In this observational study, we could not prove that pre-existing use of statins affected either the severity of the acute illness or the long-term outcome of tick-borne encephalitis.
Collapse
|
49
|
Battivelli E, Dahabieh MS, Abdel-Mohsen M, Svensson JP, Tojal Da Silva I, Cohn LB, Gramatica A, Deeks S, Greene WC, Pillai SK, Verdin E. Distinct chromatin functional states correlate with HIV latency reactivation in infected primary CD4 + T cells. eLife 2018; 7:e34655. [PMID: 29714165 PMCID: PMC5973828 DOI: 10.7554/elife.34655] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/18/2018] [Indexed: 12/21/2022] Open
Abstract
Human immunodeficiency virus (HIV) infection is currently incurable, due to the persistence of latently infected cells. The 'shock and kill' approach to a cure proposes to eliminate this reservoir via transcriptional activation of latent proviruses, enabling direct or indirect killing of infected cells. Currently available latency-reversing agents (LRAs) have however proven ineffective. To understand why, we used a novel HIV reporter strain in primary CD4+ T cells and determined which latently infected cells are reactivatable by current candidate LRAs. Remarkably, none of these agents reactivated more than 5% of cells carrying a latent provirus. Sequencing analysis of reactivatable vs. non-reactivatable populations revealed that the integration sites were distinguishable in terms of chromatin functional states. Our findings challenge the feasibility of 'shock and kill', and suggest the need to explore other strategies to control the latent HIV reservoir.
Collapse
Affiliation(s)
- Emilie Battivelli
- Gladstone Institute of Virology and ImmunologyGladstone InstitutesSan FranciscoUnited States
- Department of MedicineUniversity of California San FranciscoSan FranciscoUnited States
- Buck Institute for Research on AgingNovatoUnited States
| | - Matthew S Dahabieh
- Gladstone Institute of Virology and ImmunologyGladstone InstitutesSan FranciscoUnited States
- Department of MedicineUniversity of California San FranciscoSan FranciscoUnited States
| | - Mohamed Abdel-Mohsen
- University of California San FranciscoSan FranciscoUnited States
- Blood Systems Research InstituteSan FranciscoUnited States
- The Wistar InstitutePhiladelphiaUnited States
| | - J Peter Svensson
- Department of Biosciences and NutritionKarolinska InstitutetSolnaSweden
| | - Israel Tojal Da Silva
- Laboratory of Molecular ImmunologyThe Rockefeller UniversityNew YorkUnited States
- Laboratory of Computational Biology and BioinformaticsInternational Research CenterSao PauloBrazil
| | - Lillian B Cohn
- Laboratory of Molecular ImmunologyThe Rockefeller UniversityNew YorkUnited States
| | - Andrea Gramatica
- Gladstone Institute of Virology and ImmunologyGladstone InstitutesSan FranciscoUnited States
- Department of MedicineUniversity of California San FranciscoSan FranciscoUnited States
- Department of Cellular and Molecular PharmacologyUniversity of California San FranciscoSan FranciscoUnited States
| | - Steven Deeks
- Department of MedicineUniversity of California San FranciscoSan FranciscoUnited States
| | - Warner C Greene
- Gladstone Institute of Virology and ImmunologyGladstone InstitutesSan FranciscoUnited States
- Department of MedicineUniversity of California San FranciscoSan FranciscoUnited States
- Department of Cellular and Molecular PharmacologyUniversity of California San FranciscoSan FranciscoUnited States
| | - Satish K Pillai
- University of California San FranciscoSan FranciscoUnited States
- Blood Systems Research InstituteSan FranciscoUnited States
| | - Eric Verdin
- Gladstone Institute of Virology and ImmunologyGladstone InstitutesSan FranciscoUnited States
- Department of MedicineUniversity of California San FranciscoSan FranciscoUnited States
- Buck Institute for Research on AgingNovatoUnited States
| |
Collapse
|
50
|
Shrivastava-Ranjan P, Flint M, Bergeron É, McElroy AK, Chatterjee P, Albariño CG, Nichol ST, Spiropoulou CF. Statins Suppress Ebola Virus Infectivity by Interfering with Glycoprotein Processing. mBio 2018; 9:e00660-18. [PMID: 29717011 PMCID: PMC5930306 DOI: 10.1128/mbio.00660-18] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/18/2022] Open
Abstract
Ebola virus (EBOV) infection is a major public health concern due to high fatality rates and limited effective treatments. Statins, widely used cholesterol-lowering drugs, have pleiotropic mechanisms of action and were suggested as potential adjunct therapy for Ebola virus disease (EVD) during the 2013-2016 outbreak in West Africa. Here, we evaluated the antiviral effects of statin (lovastatin) on EBOV infection in vitro Statin treatment decreased infectious EBOV production in primary human monocyte-derived macrophages and in the hepatic cell line Huh7. Statin treatment did not interfere with viral entry, but the viral particles released from treated cells showed reduced infectivity due to inhibition of viral glycoprotein processing, as evidenced by decreased ratios of the mature glycoprotein form to precursor form. Statin-induced inhibition of infectious virus production and glycoprotein processing was reversed by exogenous mevalonate, the rate-limiting product of the cholesterol biosynthesis pathway, but not by low-density lipoprotein. Finally, statin-treated cells produced EBOV particles devoid of the surface glycoproteins required for virus infectivity. Our findings demonstrate that statin treatment inhibits EBOV infection and suggest that the efficacy of statin treatment should be evaluated in appropriate animal models of EVD.IMPORTANCE Treatments targeting Ebola virus disease (EVD) are experimental, expensive, and scarce. Statins are inexpensive generic drugs that have been used for many years for the treatment of hypercholesterolemia and have a favorable safety profile. Here, we show the antiviral effects of statins on infectious Ebola virus (EBOV) production. Our study reveals a novel molecular mechanism in which statin regulates EBOV particle infectivity by preventing glycoprotein processing and incorporation into virus particles. Additionally, statins have anti-inflammatory and immunomodulatory effects. Since inflammation and dysregulation of the immune system are characteristic features of EVD, statins could be explored as part of EVD therapeutics.
Collapse
Affiliation(s)
- Punya Shrivastava-Ranjan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mike Flint
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Anita K McElroy
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Payel Chatterjee
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - César G Albariño
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|