1
|
Chitsombat K, Chimnaronk S, Sa-ngiamsuntorn K, Chomnawang MT, Thirapanmethee K. Advancing high-throughput anti-HCV drug screening: a novel dual-reporter HCV replicon model with real-time monitoring. Res Pharm Sci 2025; 20:41-54. [PMID: 40190821 PMCID: PMC11972025 DOI: 10.4103/rps.rps_249_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/18/2024] [Accepted: 06/01/2024] [Indexed: 04/09/2025] Open
Abstract
Background and purpose Hepatitis C virus (HCV) infection is a global health concern due to its substantial impact on morbidity and mortality. The burden of diseases related to HCV continues to escalate, particularly as infections progress to late-stage liver conditions, resulting in hepatocellular carcinoma on a global scale. Direct-acting antivirals effectively target HCV replication; however, their unreasonable costs and adverse effects emphasize the need for accessible and efficient therapeutic alternatives with minimal side effects. The primary aim of this study was to devise an HCV replicon system featuring a dual-reporter mechanism to facilitate high-throughput screening of potential novel antiviral agents. Experimental approach The full-length HCV genome (pJFH1) was used to construct an HCV replicon system. The glycoprotein regions (E1 and E2) were substituted with a red fluorescent reporter, mCherry, enabling visualization of protein synthesis within the replicon. In addition, an adjacent green fluorescent reporter, dBroccoli, was strategically introduced in proximity to the NS5B stop codon to serve as a reliable indicator of HCV replication activity by monitoring the fluorescence signals. Findings/Results The findings of this study unequivocally validated the effectiveness of the novel HCV replicon system for transfecting Huh-7 cells. Furthermore, the replicon system demonstrated a concentration-dependent response to anti-HCV pharmaceutical agents including telaprevir and sofosbuvir. Conclusion and implications These compelling results underscored the potential utility of the proposed HCV replicon system as an innovative model for the expeditious high-throughput screening of prospective anti-HCV agents within a short timeframe.
Collapse
Affiliation(s)
- Kanokwan Chitsombat
- Biopharmaceutical Sciences Program, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Sarin Chimnaronk
- Institute of Molecular Biosciences, Mahidol University, Salaya Campus, Nakhon Pathom, Thailand
| | | | - Mullika Traidej Chomnawang
- Antimicrobial Resistance Interdisciplinary Center (AmRIC), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
- Department of Microbiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Krit Thirapanmethee
- Antimicrobial Resistance Interdisciplinary Center (AmRIC), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
- Department of Microbiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Prasad S, Pandey VK, Singh K, Shams R, Singh R, Goksen G. A comprehensive review on nutritional interventions and nutritive elements: Strengthening immunity for effective defense mechanism during pandemic. Food Sci Nutr 2024; 12:4534-4545. [PMID: 39055200 PMCID: PMC11266904 DOI: 10.1002/fsn3.4138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/06/2024] [Accepted: 03/16/2024] [Indexed: 07/27/2024] Open
Abstract
The pandemic has brought attention to the importance of a healthy immune system in preventing infectious diseases. In this in-depth review, the process by which nutritional interventions and fundamental nutrients affect immune function has been discussed with the goal of enhancing the body's natural defenses against viral infections. We explored the complex interplay between diet and immunology, highlighting the essential nutrients, vitamins, minerals, and bioactive substances that are crucial for enhancing immune response. We also investigated the effect of dietary patterns and supplementation methods on immune function. We assessed the effectiveness and potential mechanisms of action of various nutritional therapies in modifying immune responses through a thorough examination of scientific literature. Additionally, we go through the significance of individualized nutrition and highlight possible factors to consider for vulnerable groups, such as the elderly and people with chronic conditions. This review attempts to provide a thorough understanding of the role of diet in boosting immunity by synthesizing available research. It also offers insights into practical methods for enhancing the immune function during the current epidemic and in the future.
Collapse
Affiliation(s)
| | - Vinay Kumar Pandey
- RDC, Biotechnology DepartmentManav Rachna International Institute of Research and Studies (Deemed to Be University)FaridabadIndia
| | - Kunal Singh
- Institute of Bio Science and TechnologyShri Ramswaroop Memorial UniversityLucknowIndia
| | - Rafeeya Shams
- Department of Food Technology and NutritionLovely Professional UniversityPhagwaraIndia
| | - Rahul Singh
- Department of BioengineeringIntegral UniversityLucknowIndia
| | - Gulden Goksen
- Department of Food Technology, Vocational School of Technical Sciences at Mersin Tarsus Organized Industrial ZoneTarsus UniversityMersinTurkey
| |
Collapse
|
3
|
Méndez-Sánchez N, Coronel-Castillo CE, Ramírez-Mejía MM. Chronic Hepatitis C Virus Infection, Extrahepatic Disease and the Impact of New Direct-Acting Antivirals. Pathogens 2024; 13:339. [PMID: 38668294 PMCID: PMC11053783 DOI: 10.3390/pathogens13040339] [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: 03/06/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 04/29/2024] Open
Abstract
Chronic hepatitis C virus infection is an important cause of liver cirrhosis, hepatocellular carcinoma and death. Furthermore, it is estimated that about 40-70% of patients develop non-hepatic alterations in the course of chronic infection. Such manifestations can be immune-related conditions, lymphoproliferative disorders and metabolic alterations with serious adverse events in the short and long term. The introduction of new Direct-Acting Antivirals has shown promising results, with current evidence indicating an improvement and remission of these conditions after a sustained virological response.
Collapse
Affiliation(s)
- Nahum Méndez-Sánchez
- Unit Liver Research, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | | | - Mariana Michelle Ramírez-Mejía
- Unit Liver Research, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
- Plan of Combined Studies in Medicine (PECEM MD/PhD), Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| |
Collapse
|
4
|
Haddadzadeh Shoushtari M, Raji H, Borsi SH, Tavakol H, Cheraghian B, Moeinpour M. Evaluating the Therapeutic Effect of Sofosbuvir in Outpatients with COVID-19: A Randomized Clinical Trial Study. Galen Med J 2024; 13:e3035. [PMID: 39554397 PMCID: PMC11568422 DOI: 10.31661/gmj.v12i.3035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/25/2023] [Accepted: 07/14/2023] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has engendered scores of deaths worldwide. Just as the development of varying procedures during the pandemic has helped inhibit the disease, none is considered a definitive treatment protocol for this problem, as each induces some clinical complications pertinent to the disease. This study thus assessed the early use of sofosbuvir in outpatients with mild COVID-19. MATERIALS AND METHODS This randomized clinical trial study was conducted on 360 patients with mild COVID-19 infection at 17 Shahrivar Ahvaz Health Center. These patients were randomly divided into the intervention and control groups. Both the control and intervention groups received 400 mg of sofosbuvir and a placebo for seven days, respectively. After 14 days from the onset of the treatment, the duration of symptoms, the necessity of hospitalization, the mean of hospitalization duration, and mortality were assessed. RESULTS The most common symptoms in the intervention and control groups were coughs with a frequency of 46 (25.6%) and 54(30%), respectively. The two groups showed no statistically significant difference in the frequency of the first observed clinical symptom related to the disease (P=0.2). The mean days that the patients were symptomatic in the control group were 14±4.17, whereas, in the intervention group, it was 12.12±3.15 (P=0.08). The frequency of hospitalization in the control and intervention groups was 7 (3.8%) and 4 (2.22%), respectively (P=0.11). Moreover, the mean days of hospitalization in the control and the intervention groups were 4±1.1 and 3±0.8, respectively (P=0.15). In addition, the two groups had a similar frequency of hospitalization in the ICU (0) and mortality rate (0). CONCLUSION Sofosbuvir alone cannot play a significant role in the treatment of outpatients with mild COVID-19.
Collapse
Affiliation(s)
- Maryam Haddadzadeh Shoushtari
- Air Pollution and Respiratory Diseases Research Center, Ahvaz Jundishapur University
of Medical Sciences, Ahvaz, Iran
| | - Hanieh Raji
- Air Pollution and Respiratory Diseases Research Center, Ahvaz Jundishapur University
of Medical Sciences, Ahvaz, Iran
| | - Seyed Hamid Borsi
- Air Pollution and Respiratory Diseases Research Center, Ahvaz Jundishapur University
of Medical Sciences, Ahvaz, Iran
| | - Heshmatollah Tavakol
- Air Pollution and Respiratory Diseases Research Center, Ahvaz Jundishapur University
of Medical Sciences, Ahvaz, Iran
| | - Bahman Cheraghian
- Department of Biostatics and Public Health, Ahvaz Jundishapur University of Medical
Sciences, Ahvaz, Iran
| | - Mahtab Moeinpour
- Department of Pulmonology, School of Medicine, Ahvaz Jundishapur University of
Medical Sciences, Ahvaz, Iran
| |
Collapse
|
5
|
Kamal S, Shahzad A, Rehman K, Tariq K, Akash MSH, Imran M, Assiri MA. Therapeutic Intervention of Serine Protease Inhibitors against Hepatitis C Virus. Curr Med Chem 2024; 31:2052-2072. [PMID: 37855348 DOI: 10.2174/0109298673234823230921090431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 05/12/2023] [Accepted: 05/23/2023] [Indexed: 10/20/2023]
Abstract
Hepatitis C virus (HCV) is a globally prevalent and hazardous disorder that is responsible for inducing several persistent and potentially fatal liver diseases. Current treatment strategies offer limited efficacy, often accompanied by severe and debilitating adverse effects. Consequently, there is an urgent and compelling need to develop novel therapeutic interventions that can provide maximum efficacy in combating HCV while minimizing the burden of adverse effects on patients. One promising target against HCV is the NS3-4A serine protease, a complex composed of two HCV-encoded proteins. This non-covalent heterodimer is crucial in the viral life cycle and has become a primary focus for therapeutic interventions. Although peginterferon, combined with ribavirin, is commonly employed for HCV treatment, its efficacy is hampered by significant adverse effects that can profoundly impact patients' quality of life. In recent years, the development of direct-acting antiviral agents (DAAs) has emerged as a breakthrough in HCV therapy. These agents exhibit remarkable potency against the virus and have demonstrated fewer adverse effects when combined with other DAAs. However, it is important to note that there is a potential for developing resistance to DAAs due to alterations in the amino acid position of the NS3-4A protease. This emphasizes the need for ongoing research to identify strategies that can minimize the emergence of resistance and ensure long-term effectiveness. While the combination of DAAs holds promise for HCV treatment, it is crucial to consider the possibility of drug-drug interactions. These interactions may occur when different DAAs are used concurrently, potentially compromising their therapeutic efficacy. Therefore, carefully evaluating and monitoring potential drug interactions are vital to optimize treatment outcomes. In the pursuit of novel therapeutic interventions for HCV, the field of computational biology and bioinformatics has emerged as a valuable tool. These advanced technologies and methodologies enable the development and design of new drugs and therapeutic agents that exhibit maximum efficacy, reduced risk of resistance, and minimal adverse effects. By leveraging computational approaches, researchers can efficiently screen and optimize potential candidates, accelerating the discovery and development of highly effective treatments for HCV, treatments.
Collapse
Affiliation(s)
- Shagufta Kamal
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | - Asif Shahzad
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, The Women University, Multan, Pakistan
| | - Komal Tariq
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | | | - Muhammad Imran
- Research center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed Ali Assiri
- Research center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
6
|
Hsu CW, Yang WW, Hou CY, Feng IJ, Huang TY, Lee PL, Guo HR, Huang CY, Su SB. Patients with Hepatitis C Undergoing Direct-Acting Antiviral Treatment Have a Lower SARS-CoV-2 Infection Rate. Life (Basel) 2023; 13:2326. [PMID: 38137927 PMCID: PMC10745044 DOI: 10.3390/life13122326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
This study retrospectively analyzed the medical records of 602 patients with first-time positive results for the HCV nucleic acid test between 1 May 2021 and 31 March 2023, exploring the association between DAA treatment and SARS-CoV-2 infection. The results showed that 9.8% of HCV patients were co-infected with SARS-CoV-2. Gender, age, vaccination status, and HCV genotype did not significantly affect SARS-CoV-2 infection. However, patients undergoing DAA treatment showed significantly lower rates of SARS-CoV-2 infection and mortality compared to those not undergoing DAA treatment. The analysis also compared patients undergoing different DAA treatments, with Epclusa and Maviret showing superior protection against SARS-CoV-2. Furthermore, this study explored the severity and mortality of SARS-CoV-2 infection in patients undergoing and having completed DAA treatment. It revealed that patients diagnosed with COVID-19 during DAA treatment experienced only mild symptoms, and none died, suggesting a potential protective effect of DAA treatment against severe outcomes of SARS-CoV-2 infection. The findings contribute to the understanding of the interplay between HCV, DAA treatment, and SARS-CoV-2 infection, highlighting the need for continued monitoring and healthcare measures for individuals with chronic conditions during the ongoing COVID-19 pandemic.
Collapse
Affiliation(s)
- Chin-Wen Hsu
- Department of Family Medicine, Chi-Mei Medical Center, Liouying, Tainan 736402, Taiwan
| | - Wan-Wen Yang
- Department of Clinical Pathology, Chi-Mei Medical Center, Liouying, Tainan 736402, Taiwan
| | - Chia-Yi Hou
- Department of Clinical Pathology, Chi-Mei Medical Center, Liouying, Tainan 736402, Taiwan
| | - I-Jung Feng
- Institute of Precision Medicine, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
| | - Ting-Yi Huang
- Department of Hepato-Gastroenterology, Chi-Mei Medical Center, Liouying, Tainan 736402, Taiwan
| | - Pei-Lun Lee
- Department of Hepato-Gastroenterology, Chi-Mei Medical Center, Liouying, Tainan 736402, Taiwan
| | - How-Ran Guo
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan;
| | - Chien-Yuan Huang
- Division of Occupational Medicine, Chi-Mei Medical Center, Liouying, Tainan 736402, Taiwan
| | - Shih-Bin Su
- Division of Occupational Medicine, Chi-Mei Medical Center, Liouying, Tainan 736402, Taiwan
| |
Collapse
|
7
|
Ghebremeskel GG, Berhe Solomon M, Achila OO, Mengistu ST, Asmelash RF, Berhane Mesfin A, Hamida ME. Real-world treatment outcome of direct-acting antivirals and patient survival rates in chronic hepatitis C virus infection in Eritrea. Sci Rep 2023; 13:20792. [PMID: 38012181 PMCID: PMC10682448 DOI: 10.1038/s41598-023-47258-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/10/2023] [Indexed: 11/29/2023] Open
Abstract
Reliable real-world data on direct acting anti-retroviral (DAA) uptake and treatment outcomes are lacking for patients with hepatitis C virus (HCV) in sub-Saharan Africa. This study provides data on HCV DAA-based treatment outcomes, mortality, loss-to-follow up, and associated factors among patients in Eritrea. A multicenter retrospective observational cohort study was conducted in two tertiary hospitals in Asmara, Eritrea. A structured checklist was used to collect data from patient's cards. Descriptive and inferential statistics used included means (± Standard deviation (SD), medians (Interquartile range (IQR), chi-squire (χ2), Kaplan-Meier estimates, and multivariate Cox proportional hazard models. A total of 238 patients with median age of 59 years (IQR 50-69 years) were enrolled in the study. Out of the 227 patients initiated on treatment, 125 patients had viral load measurements at 12 weeks after end of treatment (EOT) whereas 102 patients had no viral load measurements at 12 weeks EOT. Among the patients with HCV RNA data post-EOT 12, 116 (92.8%) had sustained viral response (SVR). The prevalence of death and loss-to-follow up (LTFU) were (7.5%, 95% CI 1.7-4.1) and 67 (28.1%, 95% CI 22.3-33.9) translating into an incidence of 1.1 (95% CI 0.8-1.5) per 10,000 person days. Independent predictors of LTFU included the enrollment year (2020: aHR = 2.2, 95% CI 1-4.7; p value = 0.04); Hospital (Hospital B: aHR = 2.2, 95% CI 1-4.7; p value = 0.03) and the FIB-4 score (FIB-Score < 1.45: aHR = 3.7, 95% CI 1.2-11.5; p value = 0.02). The SVR rates achieved in this cohort were high. However, high LTFU and high mortality driven largely by late presentation and suboptimal population screening/case finding, were uncovered. These challenges can be addressed by test-and-treat programs that simultaneously prioritize programmatic screening, decentralization of care, and better patient tracking in the HCV care cascade.
Collapse
Affiliation(s)
| | | | - Oliver Okoth Achila
- Unit of Clinical Laboratory Science, Orotta College of Medicine and Health Sciences (OCMHS), Asmara, Eritrea
| | | | | | - Araia Berhane Mesfin
- National Communicable Disease Control Division, Ministry of Health, Asmara, Eritrea
| | - Mohammed Elfatih Hamida
- Department of Medical Microbiology, Orotta College of Medicine and Health Sciences (OCMHS), Asmara, Eritrea
| |
Collapse
|
8
|
Medina C, García AH, Crespo FI, Toro FI, Mayora SJ, De Sanctis JB. A Synopsis of Hepatitis C Virus Treatments and Future Perspectives. Curr Issues Mol Biol 2023; 45:8255-8276. [PMID: 37886964 PMCID: PMC10605161 DOI: 10.3390/cimb45100521] [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: 09/28/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
Hepatitis C virus (HCV) infection is a worldwide public health problem. Chronic infection with HCV can lead to liver cirrhosis or cancer. Although some immune-competent individuals can clear the virus, others develop chronic HCV disease due to viral mutations or an impaired immune response. IFNs type I and III and the signal transduction induced by them are essential for a proper antiviral effect. Research on the viral cycle and immune escape mechanisms has formed the basis of therapeutic strategies to achieve a sustained virological response (SVR). The first therapies were based on IFNα; then, IFNα plus ribavirin (IFN-RBV); and then, pegylated-IFNα-RBV (PEGIFNα-RIV) to improve cytokine pharmacokinetics. However, the maximum SVR was 60%, and several significant side effects were observed, decreasing patients' treatment adherence. The development of direct-acting antivirals (DAAs) significantly enhanced the SVR (>90%), and the compounds were able to inhibit HCV replication without significant side effects, even in paediatric populations. The management of coinfected HBV-HCV and HCV-HIV patients has also improved based on DAA and PEG-IFNα-RBV (HBV-HCV). CD4 cells are crucial for an effective antiviral response. The IFNλ3, IL28B, TNF-α, IL-10, TLR-3, and TLR-9 gene polymorphisms are involved in viral clearance, therapeutic responses, and hepatic pathologies. Future research should focus on searching for strategies to circumvent resistance-associated substitution (RAS) to DAAs, develop new therapeutic schemes for different medical conditions, including organ transplant, and develop vaccines for long-lasting cellular and humoral responses with cross-protection against different HCV genotypes. The goal is to minimise the probability of HCV infection, HCV chronicity and hepatic carcinoma.
Collapse
Affiliation(s)
- Christian Medina
- Institute of Immunology Dr. Nicolás E. Bianco C., Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (C.M.); (F.I.C.); (F.I.T.); (S.J.M.)
| | - Alexis Hipólito García
- Institute of Immunology Dr. Nicolás E. Bianco C., Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (C.M.); (F.I.C.); (F.I.T.); (S.J.M.)
| | - Francis Isamarg Crespo
- Institute of Immunology Dr. Nicolás E. Bianco C., Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (C.M.); (F.I.C.); (F.I.T.); (S.J.M.)
| | - Félix Isidro Toro
- Institute of Immunology Dr. Nicolás E. Bianco C., Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (C.M.); (F.I.C.); (F.I.T.); (S.J.M.)
| | - Soriuska José Mayora
- Institute of Immunology Dr. Nicolás E. Bianco C., Faculty of Medicine, Universidad Central de Venezuela, Caracas 1040, Venezuela; (C.M.); (F.I.C.); (F.I.T.); (S.J.M.)
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, 779 00 Olomouc, Czech Republic
- The Czech Advanced Technology and Research Institute (Catrin), Palacky University, 779 00 Olomouc, Czech Republic
| |
Collapse
|
9
|
Lachhab S, El Mansouri AE, Mehdi A, Dennemont I, Neyts J, Jochmans D, Andrei G, Snoeck R, Sanghvi YS, Ait Ali M, Loiseau PM, Lazrek HB. Synthesis of new 3-acetyl-1,3,4-oxadiazolines combined with pyrimidines as antileishmanial and antiviral agents. Mol Divers 2023; 27:2147-2159. [PMID: 36251201 PMCID: PMC9573813 DOI: 10.1007/s11030-022-10548-9] [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: 06/12/2022] [Accepted: 10/07/2022] [Indexed: 11/25/2022]
Abstract
A new series of 3-acetyl-1,3,4-oxadiazoline hybrid molecules was designed and synthesized using a condensation between acyclonucleosides and substituted phenylhydrazone. All intermediates and final products were screened against Leishmania donovani, a Protozoan parasite and against three viruses SARS-CoV-2, HCMV and VZV. While no significant activity was observed against the viruses, the intermediate with 6-azatymine as thymine and 5-azathymine-3-acetyl-1,3,4-oxadiazoline hybrid exhibited a significant antileishmanial activity. The later compound was the most promising, exhibiting an IC50 value at 8.98 µM on L. donovani intramacrophage amastigotes and a moderate selectivity index value at 2.4.
Collapse
Affiliation(s)
- Saida Lachhab
- Laboratory of Biomolecular and Medicinal Chemistry, Faculty of Science Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Az-Eddine El Mansouri
- Department of Chemistry, University of the Free State, P.O. Box 339, Bloemfontein, 9300, South Africa
| | - Ahmad Mehdi
- ICGM, Université Montpellier, CNRS, ENSCM, Montpellier, France
| | - Indira Dennemont
- Antiparasite Chemotherapy, CNRS, BioCIS, Université Paris-Saclay, Chatenay-Malabry, 92290, Paris, France
| | - Johan Neyts
- Rega Institute for Medical Research, KULeuven, Louvain, Belgium
| | - Dirk Jochmans
- Rega Institute for Medical Research, KULeuven, Louvain, Belgium
| | - Graciela Andrei
- Rega Institute for Medical Research, KULeuven, Louvain, Belgium
| | - Robert Snoeck
- Rega Institute for Medical Research, KULeuven, Louvain, Belgium
| | - Yogesh S Sanghvi
- Rasayan Inc., 2802 Crystal Ridge Road, Encinitas, CA, 92024-6615, USA
| | - Mustapha Ait Ali
- Laboratory of Biomolecular and Medicinal Chemistry, Faculty of Science Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Philippe M Loiseau
- Antiparasite Chemotherapy, CNRS, BioCIS, Université Paris-Saclay, Chatenay-Malabry, 92290, Paris, France
| | - Hassan B Lazrek
- Laboratory of Biomolecular and Medicinal Chemistry, Faculty of Science Semlalia, University Cadi Ayyad, Marrakech, Morocco.
| |
Collapse
|
10
|
Charoenkwan P, Waramit S, Chumnanpuen P, Schaduangrat N, Shoombuatong W. TROLLOPE: A novel sequence-based stacked approach for the accelerated discovery of linear T-cell epitopes of hepatitis C virus. PLoS One 2023; 18:e0290538. [PMID: 37624802 PMCID: PMC10456195 DOI: 10.1371/journal.pone.0290538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Hepatitis C virus (HCV) infection is a concerning health issue that causes chronic liver diseases. Despite many successful therapeutic outcomes, no effective HCV vaccines are currently available. Focusing on T cell activity, the primary effector for HCV clearance, T cell epitopes of HCV (TCE-HCV) are considered promising elements to accelerate HCV vaccine efficacy. Thus, accurate and rapid identification of TCE-HCVs is recommended to obtain more efficient therapy for chronic HCV infection. In this study, a novel sequence-based stacked approach, termed TROLLOPE, is proposed to accurately identify TCE-HCVs from sequence information. Specifically, we employed 12 different sequence-based feature descriptors from heterogeneous perspectives, such as physicochemical properties, composition-transition-distribution information and composition information. These descriptors were used in cooperation with 12 popular machine learning (ML) algorithms to create 144 base-classifiers. To maximize the utility of these base-classifiers, we used a feature selection strategy to determine a collection of potential base-classifiers and integrated them to develop the meta-classifier. Comprehensive experiments based on both cross-validation and independent tests demonstrated the superior predictive performance of TROLLOPE compared with conventional ML classifiers, with cross-validation and independent test accuracies of 0.745 and 0.747, respectively. Finally, a user-friendly online web server of TROLLOPE (http://pmlabqsar.pythonanywhere.com/TROLLOPE) has been developed to serve research efforts in the large-scale identification of potential TCE-HCVs for follow-up experimental verification.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, Thailand
| | - Sajee Waramit
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Pramote Chumnanpuen
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, Thailand
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, Thailand
| | - Nalini Schaduangrat
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Watshara Shoombuatong
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
11
|
Gordan JD, Keenan BP, Lim HC, Yarchoan M, Kelley RK. New Opportunities to Individualize Frontline Therapy in Advanced Stages of Hepatocellular Carcinoma. Drugs 2023; 83:1091-1109. [PMID: 37402062 DOI: 10.1007/s40265-023-01907-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2023] [Indexed: 07/05/2023]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer death globally and is rising in incidence. Until recently, treatment options for patients with advanced stages of HCC have been limited to antiangiogenic therapies with modest improvements in overall survival. The emerging role of immunotherapy with immune checkpoint inhibitors (ICI) in oncology has led to a rapid expansion in treatment options and improvements in outcomes for patients with advanced stages of HCC. Recent clinical trials have shown meaningful survival improvement in patients treated with the combination of bevacizumab and atezolizumab, as well as with the combination of tremelimumab with durvalumab, resulting in regulatory approvals of these regimens as frontline therapy. Beyond improvements in overall survival, ICI-based combination regimens achieve higher rates of durable treatment response than multikinase inhibitors and have favorable side effect profiles. With the emergence of doublet anti-angiogenic and immune checkpoint inhibitor (ICI) and dual ICI combinations, individualized therapy is now possible for patients based on co-morbidity profiles and other factors. These more potent systemic therapies are also being tested in earlier stages of disease and in combination with loco-regional therapies such as trans-arterial chemoembolization and stereotactic body radiotherapy. We summarize these advances and emerging therapeutic combinations currently in clinical trials.
Collapse
Affiliation(s)
- John D Gordan
- Division of Hematology/Oncology, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA.
- Quantitative Biosciences Institute, UC San Francisco, San Francisco, CA, USA.
| | - Bridget P Keenan
- Division of Hematology/Oncology, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA
- Cancer Immunotherapy Program, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA
| | - Huat Chye Lim
- Division of Hematology/Oncology, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute, UC San Francisco, San Francisco, CA, USA
| | - Mark Yarchoan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - R Katie Kelley
- Division of Hematology/Oncology, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA
- Cancer Immunotherapy Program, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA
| |
Collapse
|
12
|
McDuffie D, Barr D, Helm M, Baumert T, Agarwal A, Thomas E. Physiomimetic In Vitro Human Models for Viral Infection in the Liver. Semin Liver Dis 2023; 43:31-49. [PMID: 36402129 PMCID: PMC10005888 DOI: 10.1055/a-1981-5944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Viral hepatitis is a leading cause of liver morbidity and mortality globally. The mechanisms underlying acute infection and clearance, versus the development of chronic infection, are poorly understood. In vitro models of viral hepatitis circumvent the high costs and ethical considerations of animal models, which also translate poorly to studying the human-specific hepatitis viruses. However, significant challenges are associated with modeling long-term infection in vitro. Differentiated hepatocytes are best able to sustain chronic viral hepatitis infection, but standard two-dimensional models are limited because they fail to mimic the architecture and cellular microenvironment of the liver, and cannot maintain a differentiated hepatocyte phenotype over extended periods. Alternatively, physiomimetic models facilitate important interactions between hepatocytes and their microenvironment by incorporating liver-specific environmental factors such as three-dimensional ECM interactions and co-culture with non-parenchymal cells. These physiologically relevant interactions help maintain a functional hepatocyte phenotype that is critical for sustaining viral hepatitis infection. In this review, we provide an overview of distinct, novel, and innovative in vitro liver models and discuss their functionality and relevance in modeling viral hepatitis. These platforms may provide novel insight into mechanisms that regulate viral clearance versus progression to chronic infections that can drive subsequent liver disease.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Madeline Helm
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - Thomas Baumert
- Inserm Research Institute for Viral and Liver Diseases, University of Strasbourg, Strasbourg, France
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
13
|
Dwivedi M, Dwivedi A, Mukherjee D. An Insight into Hepatitis C Virus: In Search of Promising Drug Targets. Curr Drug Targets 2023; 24:1127-1138. [PMID: 37907492 DOI: 10.2174/0113894501265769231020031857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 11/02/2023]
Abstract
Hepatitis C Virus (HCV) is a global health concern, chronically infecting over 70 million people worldwide. HCV is a bloodborne pathogen that primarily affects the liver, and chronic HCV infection can lead to cirrhosis, liver cancer, and liver failure over time. There is an urgent need for more effective approaches to prevent and treat HCV. This review summarizes current knowledge on the virology, transmission, diagnosis, and management of HCV infection. It also provides an in-depth analysis of HCV proteins as promising targets for antiviral drug and vaccine development. Specific HCV proteins discussed as potential drug targets include the NS5B polymerase, NS3/4A protease, entry receptors like CD81, and core proteins. The implications of HCV proteins as diagnostic and prognostic biomarkers are also explored. Current direct-acting antiviral therapies are effective but have cost, genotype specificity, and resistance limitations. This review aims to synthesize essential information on HCV biology and pathogenesis to inform future research on improved preventive, diagnostic, and therapeutic strategies against this global infectious disease threat.
Collapse
Affiliation(s)
- Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomtinagar Extension, Lucknow- 226028, India
| | - Aditya Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomtinagar Extension, Lucknow- 226028, India
| | | |
Collapse
|
14
|
Viral proteases as therapeutic targets. Mol Aspects Med 2022; 88:101159. [PMID: 36459838 PMCID: PMC9706241 DOI: 10.1016/j.mam.2022.101159] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022]
Abstract
Some medically important viruses-including retroviruses, flaviviruses, coronaviruses, and herpesviruses-code for a protease, which is indispensable for viral maturation and pathogenesis. Viral protease inhibitors have become an important class of antiviral drugs. Development of the first-in-class viral protease inhibitor saquinavir, which targets HIV protease, started a new era in the treatment of chronic viral diseases. Combining several drugs that target different steps of the viral life cycle enables use of lower doses of individual drugs (and thereby reduction of potential side effects, which frequently occur during long term therapy) and reduces drug-resistance development. Currently, several HIV and HCV protease inhibitors are routinely used in clinical practice. In addition, a drug including an inhibitor of SARS-CoV-2 main protease, nirmatrelvir (co-administered with a pharmacokinetic booster ritonavir as Paxlovid®), was recently authorized for emergency use. This review summarizes the basic features of the proteases of human immunodeficiency virus (HIV), hepatitis C virus (HCV), and SARS-CoV-2 and discusses the properties of their inhibitors in clinical use, as well as development of compounds in the pipeline.
Collapse
|
15
|
McDuffie D, Barr D, Agarwal A, Thomas E. Physiologically relevant microsystems to study viral infection in the human liver. Front Microbiol 2022; 13:999366. [PMID: 36246284 PMCID: PMC9555087 DOI: 10.3389/fmicb.2022.999366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Viral hepatitis is a leading cause of liver disease and mortality. Infection can occur acutely or chronically, but the mechanisms that govern the clearance of virus or lack thereof are poorly understood and merit further investigation. Though cures for viral hepatitis have been developed, they are expensive, not readily accessible in vulnerable populations and some patients may remain at an increased risk of developing hepatocellular carcinoma (HCC) even after viral clearance. To sustain infection in vitro, hepatocytes must be fully mature and remain in a differentiated state. However, primary hepatocytes rapidly dedifferentiate in conventional 2D in vitro platforms. Physiologically relevant or physiomimetic microsystems, are increasingly popular alternatives to traditional two-dimensional (2D) monocultures for in vitro studies. Physiomimetic systems reconstruct and incorporate elements of the native cellular microenvironment to improve biologic functionality in vitro. Multiple elements contribute to these models including ancillary tissue architecture, cell co-cultures, matrix proteins, chemical gradients and mechanical forces that contribute to increased viability, longevity and physiologic function for the tissue of interest. These microsystems are used in a wide variety of applications to study biological phenomena. Here, we explore the use of physiomimetic microsystems as tools for studying viral hepatitis infection in the liver and how the design of these platforms is tailored for enhanced investigation of the viral lifecycle when compared to conventional 2D cell culture models. Although liver-based physiomimetic microsystems are typically applied in the context of drug studies, the platforms developed for drug discovery purposes offer a solid foundation to support studies on viral hepatitis. Physiomimetic platforms may help prolong hepatocyte functionality in order to sustain chronic viral hepatitis infection in vitro for studying virus-host interactions for prolonged periods.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
16
|
El Mansouri AE, Lachhab S, Oubella A, Ahmad M, Neyts J, Jochmans D, Chiu W, Vangeel L, De Jonghe S, Morjani H, Ali MA, Zahouily M, Sanghvi YS, Lazrek HB. Synthesis, characterization, molecular docking, and anticancer activities of new 1,3,4-oxadiazole-5-fluorocytosine hybrid derivatives. J Mol Struct 2022; 1272:134135. [PMID: 36101881 PMCID: PMC9459830 DOI: 10.1016/j.molstruc.2022.134135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 01/18/2023]
Abstract
Analogs of pyrimidine and 1,3,4-oxadiazole are two well established class of molecules proven as potent antiviral and anticancer agents in the pharmaceutical industry. We envisioned designing new molecules where these two heterocycles were conjugated with the goal of enhancing biological activity. In this vein, we synthesized a series of novel pyrimidine-1,3,4-oxadiazole conjugated hybrid molecules as potential anticancer and antiviral agents. Herein, we present a new design for 5-fluorocytosine-1,3,4-oxadiazole hybrids (5a-h) connected via a methylene bridge. An efficient synthesis of new derivatives was established, and all compounds were fully characterized by NMR and MS. Eight compounds were evaluated for their cytotoxic activity against fibrosarcoma (HT-1080), breast (MCF-7 and MDA-MB-231), lung carcinoma (A-549), and for their antiviral activity against SARS-CoV-2. Among all compounds tested, the compound 5e showed marked growth inhibition against all cell lines tested, particularly in HT-1080, with IC50 values of 19.56 µM. Meanwhile, all tested compounds showed no anti-SARS-CoV-2 activity, with EC50 >100 µM. The mechanism of cell death was investigated using Annexin V staining, caspase-3/7 activity, and analysis of cell cycle progression. The compound 5e induced apoptosis by the activation of caspase-3/7 and cell-cycle arrest in HT-1080 and A-549 cells at the G2M phase. The molecular docking suggested that the compound 5e activated caspase-3 via the formation of a stable complex protein-ligand.
Collapse
Affiliation(s)
- Az-Eddine El Mansouri
- Laboratory of Biomolecular and Medicinal chemistry, Faculty of Science Semlalia, University Cadi Ayyad, Marrakesh, Morocco
- Laboratoire de Matériaux, Catalyse & Valorisation des Ressources Naturelles, URAC 24, Faculté des Sciences et Techniques, Université Hassan II, Casablanca B.P. 146, 20650, Morocco
| | - Saida Lachhab
- Laboratory of Biomolecular and Medicinal chemistry, Faculty of Science Semlalia, University Cadi Ayyad, Marrakesh, Morocco
| | - Ali Oubella
- Laboratoire de Synthese Organique et de Physico-Chimie Moleculaire, Departement de Chimie, Faculte´ des Sciences, Semlalia BP 2390, Marrakech 40001, Morocco
| | - Mehdi Ahmad
- ICGM, Univ. Montpellier, CNRS, ENSCM, Montpellier, France
| | - Johan Neyts
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Dirk Jochmans
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Winston Chiu
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Laura Vangeel
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | - Hamid Morjani
- BioSpecT - EA7506 UFR de Pharmacie, Univ-Reims 51, rue Cognacq Jay 51096 Reims cedex, France
| | - Mustapha Ait Ali
- Laboratory of Biomolecular and Medicinal chemistry, Faculty of Science Semlalia, University Cadi Ayyad, Marrakesh, Morocco
| | - Mohamed Zahouily
- Laboratoire de Matériaux, Catalyse & Valorisation des Ressources Naturelles, URAC 24, Faculté des Sciences et Techniques, Université Hassan II, Casablanca B.P. 146, 20650, Morocco
| | - Yogesh S Sanghvi
- Rasayan Inc. 2802 Crystal Ridge Road, Encinitas, CA 92024-6615, U.S.A
| | - Hassan B Lazrek
- Laboratory of Biomolecular and Medicinal chemistry, Faculty of Science Semlalia, University Cadi Ayyad, Marrakesh, Morocco
| |
Collapse
|
17
|
Kim H, Yi J, Yu J, Park J, Jang SK. A Simple and Effective Method to Concentrate Hepatitis C Virus: Aqueous Two-Phase System Allows Highly Efficient Enrichment of Enveloped Viruses. Viruses 2022; 14:v14091987. [PMID: 36146792 PMCID: PMC9503063 DOI: 10.3390/v14091987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/05/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
To investigate the proliferation cycle of a virus, virus-host interaction, and pathogenesis of a virus, virion particles must be concentrated from the media of virus cell culture or the sera of virus-infected patients. Ultracentrifugation of the culture media is a standard method for concentrating virion particles. However, this method is time-consuming and requires special equipment (ultracentrifuge). Moreover, a large number of infectious viruses are lost during enrichment. We developed a new method of hepatitis C virus (HCV) concentration to overcome the issues associated with traditional methods of virus concentration. We used an aqueous two-phase system (ATPS) to concentrate the virus. HCV, which causes various liver diseases, such as liver fibrosis, cirrhosis, and hepatocellular carcinoma, was used as a model virus to test the efficacy and reliability of the ATPS. The efficiency of HCV concentration by the ATPS was approximately three times higher than that by ultracentrifugation. Moreover, the infectivity of the concentrated HCV, which is a labile virus, remained the same after concentration of the virus by the ATPS. Considering the simplicity and effectiveness of the ATPS, it is the method of choice for concentrating viruses.
Collapse
Affiliation(s)
- Heesun Kim
- Molecular Virology Laboratory, POSTECH Biotech Center, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Johan Yi
- Nanoparticle and Vesicle Laboratory, Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Jinbae Yu
- Molecular Virology Laboratory, Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Jaesung Park
- Nanoparticle and Vesicle Laboratory, Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang 37673, Korea
- Nanoparticle and Vesicle Laboratory, School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 37673, Korea
- Correspondence: (J.P.); (S.K.J.)
| | - Sung Key Jang
- Molecular Virology Laboratory, POSTECH Biotech Center, Pohang University of Science and Technology, Pohang 37673, Korea
- Molecular Virology Laboratory, Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
- Correspondence: (J.P.); (S.K.J.)
| |
Collapse
|
18
|
Chozie NA, Satiti MA, Sjarif DR, Oswari H, Ritchie NK. The impact of nucleic acid testing as a blood donor screening method in transfusion-associated hepatitis C among children with bleeding disorders in Indonesia: a single-center experience. Blood Res 2022; 57:129-134. [PMID: 35620907 PMCID: PMC9242827 DOI: 10.5045/br.2022.2021219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/08/2022] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Background Children with bleeding disorders, such as hemophilia and von Willebrand disease (VWD), have an increased risk of acquiring transfusion-transmitted infections (TTI). Screening methods to exclude blood donations that are at risk of transmitting infection from donors to recipients are critical to preventing disease transmission. Nucleic acid testing (NAT) is the latest blood donor-screening method. This study aimed to determine the incidence of hepatitis C virus (HCV) infection in children with hemophilia and VWD at Dr. Cipto Mangunkusumo Hospital with a history of blood transfusion before and after implementation of a NAT screening method. Methods A cohort retrospective study was conducted on children aged 0‒18 years with bleeding disorders and a history of blood transfusion. In our center, all blood transfusions before 2015 were screened using non-NAT methods, while all blood transfusions were screened using NAT starting in 2015. Eligible patient characteristics were collected from medical records. From July to December 2019, blood samples were obtained from eligible patients for anti-HCV examination. HCV RNA examinations were performed on subjects with reactive anti-HCV results, and the relative risk was calculated. Results In total, 108 eligible participants were included in this study. We observed that 91 (94.3%) patients had history of receiving non-NAT blood transfusions, while 17 (15.7%) patients received NAT-screened blood transfusions. The proportion of anti-HCV reactivity in the non-NAT group and that in the NAT group were 3.3% (3/91) and 0% (0/17), respectively. Conclusion None of the patients exhibited reactivity to anti-HCV after implementing the NAT screening method.
Collapse
Affiliation(s)
- Novie Amelia Chozie
- Department of Child Health, Faculty of Medicine University of Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta Pusat, Indonesia
| | - Melati Arum Satiti
- Department of Child Health, Faculty of Medicine University of Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta Pusat, Indonesia
| | - Damayanti Rusli Sjarif
- Department of Child Health, Faculty of Medicine University of Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta Pusat, Indonesia
| | - Hanifah Oswari
- Department of Child Health, Faculty of Medicine University of Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta Pusat, Indonesia
| | - Ni Ken Ritchie
- Jakarta Blood Center, Indonesian Red Cross Society, Jakarta Pusat, Indonesia
| |
Collapse
|
19
|
Mawuli G, Dzudzor B, Tachi K, Kuma AABA, Odame-Aboagye J, Obeng BM, Boateng AT, Edu-Quansah EP, Attiku KO, Agbosu E, Arjarquah A, Bonney JHK. Hepatitis C virus (HCV) infection among patients with sickle cell disease at the Korle-Bu teaching hospital. Virol J 2022; 19:73. [PMID: 35459145 PMCID: PMC9026067 DOI: 10.1186/s12985-022-01797-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/11/2022] [Indexed: 11/12/2022] Open
Abstract
Background Hepatitis C virus (HCV) infection is a blood borne infection that remains potentially transmissible through blood transfusions. Sickle cell disease (SCD) is a common inheritable haemoglobinopathy in Ghana that requires multiple blood transfusions as part of its management. The SCD patient is therefore at a high risk of HCV infection; however, data on the occurrence of HCV in SCD patients has not been documented in Ghana. This study sought to determine the prevalence and genotypes of HCV infection in SCD patients. Materials and methods This was a cross-sectional study which enrolled 141 sickle-cell disease patients from the Ghana Institute for Clinical Genetics, Korle-Bu Teaching Hospital (KBTH). Patient information was obtained through a structured questionnaire. Aliquots of the plasma obtained was used for both serology with Advanced Quality Rapid Anti-HCV Test Strip and molecular testing by RT-PCR with primers targeting the HCV core gene. The amplified DNA were purified and subjected to phylogenetic analysis to characterize HCV genotypes. Results Twelve (9%) out of the 141 patients were sero-positive for HCV total antibodies. HCV RNA was amplified from 8 (6%) out of the total number of patients’ samples. One of the 12 sero-positives was HCV RNA positive. Five (63%) out of the 8 HCV RNA positive samples were successfully sequenced. The phylogenetic tree constructed with the study and GenBank reference sequences, clustered all five study sequences into HCV genotype 1. Conclusion The HCV seroprevalence of 9% among sickle cell disease patients is higher than reported for the general Ghanaian population which is 3%. Genotype 1 is the common HCV genotype infecting SCD patients. Sickle cell disease is likely to be a high-risk group for HCV inapparent infections in Ghana as seroprevalence does not correlate with viremia. However, even with higher seroprevalence, the group must be given priority in resource allocation for preventive, diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Gifty Mawuli
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, P. O. Box, LG 581, Legon Accra, Ghana.,Department of Medical Biochemistry, University of Ghana Medical School, University of Ghana, Accra, Ghana
| | - Bartholomew Dzudzor
- Department of Medical Biochemistry, University of Ghana Medical School, University of Ghana, Accra, Ghana
| | - Kenneth Tachi
- Department of Medicine and Therapeutics, University of Ghana Medical School, University of Ghana, Accra, Ghana
| | | | - James Odame-Aboagye
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, P. O. Box, LG 581, Legon Accra, Ghana
| | | | - Anthony Twumasi Boateng
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, P. O. Box, LG 581, Legon Accra, Ghana
| | | | - Keren Okyerebea Attiku
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, P. O. Box, LG 581, Legon Accra, Ghana
| | - Esinam Agbosu
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, P. O. Box, LG 581, Legon Accra, Ghana
| | - Augustina Arjarquah
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, P. O. Box, LG 581, Legon Accra, Ghana
| | - Joseph Humphrey Kofi Bonney
- Department of Virology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, P. O. Box, LG 581, Legon Accra, Ghana.
| |
Collapse
|
20
|
Kamal A, Elsheaita A, Abdelnabi M. Association between direct-acting antiviral agents in hepatitis C virus treatment and hepatocellular carcinoma occurrence and recurrence: The endless debate. World J Clin Cases 2022; 10:1764-1774. [PMID: 35317156 PMCID: PMC8891795 DOI: 10.12998/wjcc.v10.i6.1764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/05/2021] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
Since direct-acting antiviral agents (DAAs) have been introduced into hepatitis C virus treatment, the sustained viral response (SVR) rate has significantly increased to more than 95%. Scientific evidence supports the idea that SVR after interferon therapy has beneficial effects related to cirrhosis progression, resulting in a reduction in the incidence of hepatocellular carcinoma (HCC). However, a significant debate exists related to DAA impact on HCC development. We reviewed the current literature highlighting the controversial data related to DAA association with de novo HCC occurrence or recurrence and possible pathophysiology of HCC related to DAAs. After a review of the published literature, we believe that the current evidence does not confirm or repudiate a higher rate of de novo HCC occurrence or recurrence related to DAA therapy. More trials are needed to determine if there is an association between HCC occurrence or recurrence and DAA or if it is related to preexisting liver cirrhosis.
Collapse
Affiliation(s)
- Ahmed Kamal
- Internal Medicine Department, Faculty of Medicine Alexandria University, Alexandria 21131, Egypt
| | - Ahmed Elsheaita
- Clinical and Experimental Internal Medicine Department, Medical Research Institute Alexandria University, Alexandria 21561, Egypt
| | - Mahmoud Abdelnabi
- Clinical and Experimental Internal Medicine Department, Medical Research Institute Alexandria University, Alexandria 21561, Egypt
- Internal Medicine Department, Texas Tech University Health Science Center, Lubbock, TX 79430, United States
| |
Collapse
|
21
|
Torres-Vázquez B, María de Lucas A, García-Crespo C, Antonio García-Martín J, Fragoso A, Fernández-Algar M, Perales C, Domingo E, Moreno M, Briones C. In vitro selection of high affinity DNA and RNA aptamers that detect hepatitis C virus core protein of genotypes 1 to 4 and inhibit virus production in cell culture. J Mol Biol 2022; 434:167501. [PMID: 35183559 DOI: 10.1016/j.jmb.2022.167501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 02/07/2023]
|
22
|
Permanasari AA, Aoki-Utsubo C, Wahyuni TS, Tumewu L, Adianti M, Widyawaruyanti A, Hotta H, Hafid AF. An in vitro study of an Artocarpus heterophyllus substance as a hepatitis C antiviral and its combination with current anti-HCV drugs. BMC Complement Med Ther 2021; 21:260. [PMID: 34641875 PMCID: PMC8507375 DOI: 10.1186/s12906-021-03408-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Background Current therapy of chronic hepatitis C virus (HCV) with direct-acting antivirals (DAAs) has dramatically improved the sustained virologic response (SVR) of affected patients; however, treatment with DAAs remains expensive, and drug-resistant HCV variants remain a threat. As a result, there is still a need to continue to develop affordable and effective drugs for the treatment of HCV. Previously, we have demonstrated that a crude extract from Artocarpus heterophyllus leaves is a potential anti-HCV candidate. In this study, we have further purified this crude extract, examined which sub-fraction possesses the highest antiviral activity, and then explored its efficacy at different HCV life cycle stages. We also assessed synergistic antiviral effects between the A. heterophyllus extract and commercially available anti-HCV drugs. Methods We used vacuum liquid chromatography (VLC) and high-performance liquid chromatography (HPLC) to fractionate a dichloromethane extract of A. heterophyllus leaves. We then examined the anti-HCV activity of the fractions using HCV genotype 2a, JFH1a; the antiviral mode of action was determined by exploring adding the treatments at different times. We examined the antiviral effects on the viral entry stage through a virucidal activity test, viral adsorption examination, and pretreatment of cells with the drug. The effects on the post-viral entry stage were determined by the levels of HCV protein expression and HCV RNA expression in infected cells. Results Through activity guided purification, we identified the sub-fraction FR3T3 as possessing the most robust anti-HCV activity with an IC50 value of 4.7 ± 1.0 μg/mL. Mode-of-action analysis revealed that FR3T3 inhibited post-viral entry stages such as HCV NS3 protein expression and HCV RNA replication with marginal effects on the viral entry stage. Thin-layer Chromatography (TLC) indicated that FR3T3 contained terpenoids and chlorophyll-related compounds. We also found a synergistic antiviral activity when the DCM extract of A. heterohyllus was used in combination therapy with commercial anti-HCV drugs; Ribavirin, Simeprevir, Cyclosporin A. Conclusions The extract of A. heterophyllus and its sub-fraction, FR3T3, presented here have anti-HCV activities and could be candidate drugs for add-on-therapy for treatment of chronic HCV infections.
Collapse
Affiliation(s)
| | - Chie Aoki-Utsubo
- Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Tutik Sri Wahyuni
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, 60115, Indonesia.,Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Lidya Tumewu
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Myrna Adianti
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, 60115, Indonesia.,Department of Health, Study Program Traditional Medicine, Vocational Faculty, Universitas Airlangga, Surabaya, Indonesia
| | - Aty Widyawaruyanti
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, 60115, Indonesia.,Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Hak Hotta
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, 6-2-23, Morikita-machi, Higashida-ku, Kobe, 658-0001, Japan
| | - Achmad Fuad Hafid
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, 60115, Indonesia. .,Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, 60115, Indonesia.
| |
Collapse
|
23
|
Sudhindar PD, Wainwright D, Saha S, Howarth R, McCain M, Bury Y, Saha SS, McPherson S, Reeves H, Patel AH, Faulkner GJ, Lunec J, Shukla R. HCV Activates Somatic L1 Retrotransposition-A Potential Hepatocarcinogenesis Pathway. Cancers (Basel) 2021; 13:5079. [PMID: 34680227 PMCID: PMC8533982 DOI: 10.3390/cancers13205079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/29/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatitis C virus (HCV) is a common cause of hepatocellular carcinoma (HCC). The activation and mutagenic consequences of L1 retrotransposons in virus-associated-HCC have been documented. However, the direct influence of HCV upon L1 elements is unclear, and is the focus of the present study. L1 transcript expression was evaluated in a publicly available liver tissue RNA-seq dataset from patients with chronic HCV hepatitis (CHC), as well as healthy controls. L1 transcript expression was significantly higher in CHC than in controls. L1orf1p (a L1 encoded protein) expression was observed in six out of 11 CHC livers by immunohistochemistry. To evaluate the influence of HCV on retrotransposition efficiency, in vitro engineered-L1 retrotransposition assays were employed in Huh7 cells in the presence and absence of an HCV replicon. An increased retrotransposition rate was observed in the presence of replicating HCV RNA, and persisted in cells after viral clearance due to sofosbuvir (PSI7977) treatment. Increased retrotransposition could be due to dysregulation of the DNA-damage repair response, including homologous recombination, due to HCV infection. Altogether these data suggest that L1 expression can be activated before oncogenic transformation in CHC patients, with HCV-upregulated retrotransposition potentially contributing to HCC genomic instability and a risk of transformation that persists post-viral clearance.
Collapse
Affiliation(s)
- Praveen D. Sudhindar
- Newcastle University Centre for Cancer, Biosciences Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.D.S.); (D.W.); (R.H.); (J.L.)
| | - Daniel Wainwright
- Newcastle University Centre for Cancer, Biosciences Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.D.S.); (D.W.); (R.H.); (J.L.)
| | - Santu Saha
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (S.S.); (M.M.); (S.S.S.); (H.R.)
| | - Rachel Howarth
- Newcastle University Centre for Cancer, Biosciences Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.D.S.); (D.W.); (R.H.); (J.L.)
| | - Misti McCain
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (S.S.); (M.M.); (S.S.S.); (H.R.)
| | - Yvonne Bury
- Department of Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK;
| | - Sweta S. Saha
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (S.S.); (M.M.); (S.S.S.); (H.R.)
| | - Stuart McPherson
- The Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Heaton NE7 7DN, UK;
| | - Helen Reeves
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (S.S.); (M.M.); (S.S.S.); (H.R.)
- The Liver Unit, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Heaton NE7 7DN, UK;
| | - Arvind H. Patel
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G61 1QH, UK;
| | - Geoffrey J. Faulkner
- Mater Research Institute, University of Queensland, Woolloongabba, QLD 4102, Australia;
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - John Lunec
- Newcastle University Centre for Cancer, Biosciences Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.D.S.); (D.W.); (R.H.); (J.L.)
| | - Ruchi Shukla
- Newcastle University Centre for Cancer, Biosciences Institute, Faculty of Medical Sciences, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (P.D.S.); (D.W.); (R.H.); (J.L.)
| |
Collapse
|
24
|
Dowarah J, Marak BN, Yadav UCS, Singh VP. Potential drug development and therapeutic approaches for clinical intervention in COVID-19. Bioorg Chem 2021; 114:105016. [PMID: 34144277 PMCID: PMC8143914 DOI: 10.1016/j.bioorg.2021.105016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/20/2021] [Indexed: 01/25/2023]
Abstract
While the vaccination is now available to many countries and will slowly dissipate to others, effective therapeutics for COVID-19 is still illusive. The SARS-CoV-2 pandemic has posed an unprecedented challenge to researchers, scientists, and clinicians and affected the wellbeing of millions of people worldwide. Since the beginning of the pandemic, a multitude of existing anti-viral, antibiotic, antimalarial, and anticancer drugs have been tested, and some have shown potency in the treatment and management of COVID-19, albeit others failed to leave any positive impact and a few also became controversial as they showed mixed clinical outcomes. In the present article, we have brought together some of the candidate therapeutic drugs being repurposed or used in the clinical trials and discussed their clinical efficacy and safety for COVID-19.
Collapse
Affiliation(s)
- Jayanta Dowarah
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | - Brilliant N Marak
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | | | - Ved Prakash Singh
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India; Department of Industrial Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India.
| |
Collapse
|
25
|
Kowalczyk M, Golonko A, Świsłocka R, Kalinowska M, Parcheta M, Swiergiel A, Lewandowski W. Drug Design Strategies for the Treatment of Viral Disease. Plant Phenolic Compounds and Their Derivatives. Front Pharmacol 2021; 12:709104. [PMID: 34393787 PMCID: PMC8363300 DOI: 10.3389/fphar.2021.709104] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus pandemic (SARS CoV-2) that has existed for over a year, constantly forces scientists to search for drugs against this virus. In silico research and selected experimental data have shown that compounds of natural origin such as phenolic acids and flavonoids have promising antiviral potential. Phenolic compounds inhibit multiplication of viruses at various stages of the viral life cycle, e.g., attachment (disturbance of the interaction between cellular and viral receptors), penetration (inhibition of viral pseudo-particle fusion to the host membrane), replication (inhibition of integrase and 3C-like protease), assembly and maturation (inhibition of microsomal triglyceride transfer protein (MTP) activity hydrolysis) and release (inhibition of secretion of apolipoprotein B (apoB) from infected cells). Phenolic compounds also indirectly influence on the viral life cycle by affecting the host cell's biochemical processes that viruses use for their own benefit. Phenolic compounds may inhibit the proteasomes and cellular deubiquitinating activity that causes an increase in the ubiquitinated proteins level in host cells. This, in turn, contributes to the lowering the available ubiquitin molecules that viruses could use for their own replication. One of the drug design strategy for the treatment of viral diseases may be an enhancement of the antiviral properties of phenolic compounds by metal complexation. Many studies have shown that the presence of a metal ion in the structure can significantly affect the affinity of the compound to key structural elements of the SARS CoV-2, such as Mpro protease, RNA-dependent RNA polymerase (RdRp) and spike protein. We believe that in the era of coronavirus pandemic, it is necessary to reconsider the search for therapeutics among well-known compounds of plant origin and their metal complexes.
Collapse
Affiliation(s)
- Monika Kowalczyk
- Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Institute of Agricultural and Food Biotechnology—State Research Institute, Warsaw, Poland
| | - Aleksandra Golonko
- Department of Microbiology, Institute of Agricultural and Food Biotechnology—State Research Institute, Warsaw, Poland
| | - Renata Świsłocka
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Bialystok, Poland
| | - Monika Kalinowska
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Bialystok, Poland
| | - Monika Parcheta
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Bialystok, Poland
| | - Artur Swiergiel
- Faculty of Biology, University of Gdansk, Gdansk, Poland
- Institute of Agricultural and Food Biotechnology—State Research Institute, Warsaw, Poland
| | | |
Collapse
|
26
|
Kardani K, Sadat SM, Kardani M, Bolhassani A. The next generation of HCV vaccines: a focus on novel adjuvant development. Expert Rev Vaccines 2021; 20:839-855. [PMID: 34114513 DOI: 10.1080/14760584.2021.1941895] [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] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Considerable efforts have been made to treat and prevent acute and chronic infections caused by the hepatitis C virus (HCV). Current treatments are unable to protect people from reinfection. Hence, there is a need for development of both preventive and therapeutic HCV vaccines. Many vaccine candidates are in development to fight against HCV, but their efficacy has so far proven limited partly due to low immunogenicity. AREAS COVERED We explore development of novel and powerful adjuvants to achieve an effective HCV vaccine. The basis for developing strong adjuvants is to understand the innate immunity pathway, which subsequently stimulates humoral and cellular immune responses. We have also investigated immunogenicity of developed adjuvants that have been used in recent studies available in online databases such as PubMed, PMC, ScienceDirect, Google Scholar, etc. EXPERT OPINION Adjuvants are used as a part of vaccine formulation to boost vaccine immunogenicity and antigen delivery. Several FDA-approved adjuvants are used in licensed human vaccines. Unfortunately, no adjuvant has yet been proven to boost HCV immune responses to the extent needed for an effective vaccine. One of the promising approaches for developing an effective adjuvant is the combination of various adjuvants to trigger several innate immune responses, leading to activation of adaptive immunity.[Figure: see text].
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Mona Kardani
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
27
|
Singh A, Gera A, Misra A, Mehndiratta S. SARS-CoV-2 infection in a pediatric acute leukemia patient on chemotherapy and concurrent sofosbuvir/velpatasvir for HCV. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:286-289. [PMID: 34322293 PMCID: PMC8303012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/05/2021] [Indexed: 06/13/2023]
Abstract
There are new targets identified by experimental and animal research for treatment of SARS-COV-2 (Severe acute respiratory syndrome-Corona Virus-2) infection. Out of many clinical trials registered, there are ongoing human studies highlighting Sofosbuvir's possible role in the treatment of Covid-19 (Coronavirus Disease 2019). Here we present a case of acute leukemia on directly acting antiviral therapy (DAAs) for HCV infection mitigating SARS-COV-2 infection in a patient undergoing chemotherapy. The child was undergoing chemotherapy, along with directly acting antiviral for acute hepatitis C infection. He initially had features of hypoxia and radiological evidence of covid-19. He had an uneventful course and tested negative ten days after onset of illness. With ongoing trials on Sofosbuvir in covid 19 treatment, our finding, albeit coincidental, points to the possible role even in immune-compromised children.
Collapse
Affiliation(s)
- Amitabh Singh
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, VMMC and Safdarjung HospitalNew Delhi 110029, India
| | - Akriti Gera
- Department of Pediatrics, VMMC and Safdarjung HospitalNew Delhi 110029, India
| | - Aroonima Misra
- Department of Health Research, National Institute of PathologyNew Delhi 110029, India
| | - Sumit Mehndiratta
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, VMMC and Safdarjung HospitalNew Delhi 110029, India
| |
Collapse
|
28
|
Tarannum H, Chauhan B, Samadder A, Roy H, Nandi S. To Explore the Potential Targets and Current Structure-based Design Strategies Utilizing Co-crystallized Ligand to Combat HCV. Curr Drug Targets 2021; 22:590-604. [PMID: 32720601 DOI: 10.2174/1389450121999200727215020] [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: 03/06/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hepatitis C Virus (HCV) belongs to the Hepacivirus family. HCV has been designated as a very dreadful virus as it can attack the liver, causing inflammation and even may lead to cancer in chronic conditions. It was estimated that 71 million people around the world have chronic HCV infection. World Health Organization (WHO) reported that about 399000 people died because of chronic cirrhosis and liver cancer globally. In spite of the abundance of availability of drugs for the treatment of HCV, however, the issue of drug resistance surpasses all the possibilities of therapeutic management of HCV. Therefore, to address this issue of 'drug-resistance', various HCV targets were explored to quest the evaluation of the mechanism of the disease progression. METHODS An attempt has been made in the present study to explore the various targets of HCV involved in the mechanism(s) of the disease initiation and progression and to focus on the mode of binding of ligands, which are co-crystallized at the active cavity of different HCV targets. CONCLUSION The present study could predict some crucial features of these ligands, which possibly interacted with various amino acid residues responsible for their biological activity and molecular signaling pathway(s). Such binding mode may be considered as a template for the high throughput screening and designing of active congeneric ligands to combat HCV.
Collapse
Affiliation(s)
- Heena Tarannum
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur-244713, India
| | - Bhumika Chauhan
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur-244713, India
| | - Asmita Samadder
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Harekrishna Roy
- Nirmala College of Pharmacy, Mangalagiri, Guntur, Andhra Pradesh, 522503, India
| | - Sisir Nandi
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur-244713, India
| |
Collapse
|
29
|
Kasyanenko KV, Kozlov KV, Maltsev OV, Lapikov II, Gordienko VV, Sharabhanov VV, Sorokin PV, Zhdanov KV. [Evaluation of the effectiveness of Riamilovir in the complex therapy of patients with COVID-19]. TERAPEVT ARKH 2021; 93:290-294. [PMID: 36286698 DOI: 10.26442/00403660.2021.03.200719] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 11/22/2022]
Abstract
AIM In this study we evaluated the effects of Riamilovir on SARS-CoV-2 viral shedding and on admission duration in patients with moderate COVID-19. MATERIALS AND METHODS We have used data from 69 health records of patients with moderate severe PCR confirmed SARS-CoV-2 infection. Control group included 34 patients treated with off-label riamilovir 1250 mg per day for 5 days (250 mg 5 times a day), comparison groups 35 patients, who received ribavirin and umifenovir 800 mg a day for 5 days. The antiviral therapy was administered within 72 hours from the onset of the disease. The primary endpoints were elimination of virus in oropharyngeal and nasopharyngeal swabs on 7 day of admission and discharge from the hospital by 14 day. RESULTS Patients assigned to riamilovir had significantly shorter time to clinical improvement as well as increased PCR negative rate by day 7. CONCLUSION Yearly administration of riamilovir as opposed to the umifenovir and ribavirin in therapy of moderate SARS-CoV-2 infection was associated with significant shorter time to clinical improvement by 14 day of hospitalization. PCR negative rate by 7 days of hospitalization is significantly more likely in riamilovir group.
Collapse
|
30
|
Onorato L, Pisaturo M, Starace M, Minichini C, Di Fraia A, Astorri R, Coppola N. Virological Factors Associated with Failure to the Latest Generation of Direct Acting Agents (DAA) and Re-Treatment Strategy: A Narrative Review. Viruses 2021; 13:432. [PMID: 33800289 PMCID: PMC8000640 DOI: 10.3390/v13030432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
The availability of all oral direct acting antiviral agents (DAAs) has revolutionized the management of HCV infections in recent years, allowing to achieve a sustained virological response (SVR) in more than 95% of cases, irrespective of hepatitis C Virus (HCV) genotype or staging of liver disease. Although rare, the failure to the latest-generation regimens (grazoprevir/elbasvir, sofosbuvir/velpatasvir, pibrentasvir/glecaprevir) represents a serious clinical problem, since the data available in the literature on the virological characteristics and management of these patients are few. The aim of the present narrative review was to provide an overview of the impact of baseline RASs in patients treated with the latest-generation DAAs and to analyze the efficacy of the available retreatment strategies in those who have failed these regimens.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nicola Coppola
- Department of Mental Health and Public Medicine, Section of Infectious Diseases, University of Campania L. Vanvitelli, 80138 Naples, Italy; (L.O.); (M.P.); (M.S.); (C.M.); (A.D.F.); (R.A.)
| |
Collapse
|
31
|
Changes of Gut-Microbiota-Liver Axis in Hepatitis C Virus Infection. BIOLOGY 2021; 10:biology10010055. [PMID: 33451143 PMCID: PMC7828638 DOI: 10.3390/biology10010055] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/02/2021] [Accepted: 01/08/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary Gut microbiota alteration is linked to many health disorders including hepatitis C virus (HCV) infection. This dysbiosis in turn impacts the coordination between the gut and the liver that is known as the gut–liver-axis. Here, we discuss the latest findings regarding the changes in gut microbiota structure and functionality post HCV infection and its treatment regimens. In addition, we underline the contribution of the microbiota alterations to HCV associated liver complications. Abstract The gut–liver-axis is a bidirectional coordination between the gut, including microbial residents, the gut microbiota, from one side and the liver on the other side. Any disturbance in this crosstalk may lead to a disease status that impacts the functionality of both the gut and the liver. A major cause of liver disorders is hepatitis C virus (HCV) infection that has been illustrated to be associated with gut microbiota dysbiosis at different stages of the disease progression. This dysbiosis may start a cycle of inflammation and metabolic disturbance that impacts the gut and liver health and contributes to the disease progression. This review discusses the latest literature addressing this interplay between the gut microbiota and the liver in HCV infection from both directions. Additionally, we highlight the contribution of gut microbiota to the metabolism of antivirals used in HCV treatment regimens and the impact of these medications on the microbiota composition. This review sheds light on the potential of the gut microbiota manipulation as an alternative therapeutic approach to control the liver complications post HCV infection.
Collapse
|
32
|
Serum Interleukin-35 Level in Correlation with T Regulatory and T helper-17 Cells Frequency in Chronic Viral Hepatitis C Patients. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2020. [DOI: 10.22207/jpam.14.4.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin-35 (IL-35), secreted mainly by T-regulatory cells (T-regs), has been considered to have immunosuppressive actions in many auto-immune diseases and tumors. However, information about its role in chronic hepatitis C (CHC) infection is still limited. We aimed to study the role of IL-35 within CHC infection and to assess its correlation with T-regs and T-helper 17 cells (Th-17). Therefore, we measured serum IL-35 concentrations using ELISA assay in 25 normal controls (NCs) and in 30 CHC patients before receiving direct antiviral agents (DAA) treatment and after 3 months of treatment end. T-regs and Th-17 cells frequencies were assessed via flow-cytometry in control group and patients’ group before treatment. The results showed that serum IL-35 levels revealed a highly significant increase in CHC patients compared to NCs (P <0.001). Moreover, IL-35 levels significantly decreased in patients 3 months after treatment end (P =0.02). Both Th-17 and T-regs were significantly increased in patients more than in NCs and a positive correlation was observed between them. However, T-regs/Th-17 ratio did not show significant difference from the ratio in NCs. IL-35 levels were positively correlated with viral load and T-regs frequency, but not with Th-17 frequency. IL-35 levels did not correlate with liver enzymes or functions. These results suggested that IL-35 enhances the immunosuppressive functions of T-regs, protecting the liver from HCV induced damage and contributes to viral persistence. IL-35 may represent a possible immunotherapeutic strategy for chronic persistent infection if given with DAA, especially in relapsing or non-responding cases.
Collapse
|
33
|
Ju J, Li X, Kumar S, Jockusch S, Chien M, Tao C, Morozova I, Kalachikov S, Kirchdoerfer RN, Russo JJ. Nucleotide analogues as inhibitors of SARS-CoV Polymerase. Pharmacol Res Perspect 2020; 8:e00674. [PMID: 33124786 PMCID: PMC7596664 DOI: 10.1002/prp2.674] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 01/18/2023] Open
Abstract
SARS-CoV-2, a member of the coronavirus family, has caused a global public health emergency. Based on our analysis of hepatitis C virus and coronavirus replication, and the molecular structures and activities of viral inhibitors, we previously reasoned that the FDA-approved hepatitis C drug EPCLUSA (Sofosbuvir/Velpatasvir) should inhibit coronaviruses, including SARS-CoV-2. Here, using model polymerase extension experiments, we demonstrate that the active triphosphate form of Sofosbuvir is incorporated by low-fidelity polymerases and SARS-CoV RNA-dependent RNA polymerase (RdRp), and blocks further incorporation by these polymerases; the active triphosphate form of Sofosbuvir is not incorporated by a host-like high-fidelity DNA polymerase. Using the same molecular insight, we selected 3'-fluoro-3'-deoxythymidine triphosphate and 3'-azido-3'-deoxythymidine triphosphate, which are the active forms of two other anti-viral agents, Alovudine and AZT (an FDA-approved HIV/AIDS drug) for evaluation as inhibitors of SARS-CoV RdRp. We demonstrate the ability of two of these HIV reverse transcriptase inhibitors to be incorporated by SARS-CoV RdRp where they also terminate further polymerase extension. Given the 98% amino acid similarity of the SARS-CoV and SARS-CoV-2 RdRps, we expect these nucleotide analogues would also inhibit the SARS-CoV-2 polymerase. These results offer guidance to further modify these nucleotide analogues to generate more potent broad-spectrum anti-coronavirus agents.
Collapse
Affiliation(s)
- Jingyue Ju
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of Chemical EngineeringColumbia UniversityNew YorkNYUSA
- Department of Molecular Pharmacology and TherapeuticsColumbia UniversityNew YorkNYUSA
| | - Xiaoxu Li
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of Chemical EngineeringColumbia UniversityNew YorkNYUSA
| | - Shiv Kumar
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of Chemical EngineeringColumbia UniversityNew YorkNYUSA
| | - Steffen Jockusch
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of ChemistryColumbia UniversityNew YorkNYUSA
| | - Minchen Chien
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of Chemical EngineeringColumbia UniversityNew YorkNYUSA
| | - Chuanjuan Tao
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of Chemical EngineeringColumbia UniversityNew YorkNYUSA
| | - Irina Morozova
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of Chemical EngineeringColumbia UniversityNew YorkNYUSA
| | - Sergey Kalachikov
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of Chemical EngineeringColumbia UniversityNew YorkNYUSA
| | - Robert N. Kirchdoerfer
- Department of BiochemistryUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Institute of Molecular VirologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - James J. Russo
- Center for Genome Technology and Biomolecular EngineeringColumbia UniversityNew YorkNew YorkUSA
- Department of Chemical EngineeringColumbia UniversityNew YorkNYUSA
| |
Collapse
|
34
|
Laugel E, Hartard C, Jeulin H, Berger S, Venard V, Bronowicki JP, Schvoerer E. Full-length genome sequencing of RNA viruses-How the approach can enlighten us on hepatitis C and hepatitis E viruses. Rev Med Virol 2020; 31:e2197. [PMID: 34260779 DOI: 10.1002/rmv.2197] [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: 08/14/2020] [Revised: 11/04/2020] [Accepted: 11/04/2020] [Indexed: 12/09/2022]
Abstract
Among the five main viruses responsible for human hepatitis, hepatitis C virus (HCV) and hepatitis E virus (HEV) are different while sharing similarities. Both viruses can be transmitted by blood or derivatives whereas HEV can also follow environmental or zoonotic routes. These highly variable RNA viruses can cause chronic hepatitis potentially leading to hepatocarcinoma. HCV and HEV can develop new structures and functions under selective pressure to adapt to host immunity, human tissues, treatments or even various animal reservoirs. Elsewhere, with directly acting antiviral treatments, HCV can be eradicated whereas HEV is an emerging pathogen against which specific treatments have to be improved. As a unique molecular tool able to explore viral genomic plasticity, full-length genome (FLG) sequencing has become easier, faster and cheaper. The present review will show how FLG sequencing can explore these RNA viruses with the aim to investigate key genomics data to improve basic knowledge, patients' healthcare and preventive tools.
Collapse
Affiliation(s)
- Elodie Laugel
- Université de Lorraine, Vandœuvre-lès-Nancy, France.,Laboratoire de Virologie, CHRU de Nancy Brabois, Vandœuvre-lès-Nancy, France.,Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l'Environnement (LCPME), UMR 7564 CNRS-UL, Vandœuvre-lès-Nancy, France
| | - Cédric Hartard
- Université de Lorraine, Vandœuvre-lès-Nancy, France.,Laboratoire de Virologie, CHRU de Nancy Brabois, Vandœuvre-lès-Nancy, France.,Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l'Environnement (LCPME), UMR 7564 CNRS-UL, Vandœuvre-lès-Nancy, France
| | - Hélène Jeulin
- Université de Lorraine, Vandœuvre-lès-Nancy, France.,Laboratoire de Virologie, CHRU de Nancy Brabois, Vandœuvre-lès-Nancy, France.,Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l'Environnement (LCPME), UMR 7564 CNRS-UL, Vandœuvre-lès-Nancy, France
| | - Sibel Berger
- Laboratoire de Virologie, CHRU de Nancy Brabois, Vandœuvre-lès-Nancy, France
| | - Véronique Venard
- Université de Lorraine, Vandœuvre-lès-Nancy, France.,Laboratoire de Virologie, CHRU de Nancy Brabois, Vandœuvre-lès-Nancy, France
| | - Jean-Pierre Bronowicki
- Université de Lorraine, Vandœuvre-lès-Nancy, France.,Service d'hépato-gastroentérologie, CHRU de Nancy Brabois, Vandœuvre-lès-Nancy, France
| | - Evelyne Schvoerer
- Université de Lorraine, Vandœuvre-lès-Nancy, France.,Laboratoire de Virologie, CHRU de Nancy Brabois, Vandœuvre-lès-Nancy, France.,Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l'Environnement (LCPME), UMR 7564 CNRS-UL, Vandœuvre-lès-Nancy, France
| |
Collapse
|
35
|
Abo-Zeid Y, Williams GR, Touabi L, McLean GR. An investigation of rhinovirus infection on cellular uptake of poly (glycerol-adipate) nanoparticles. Int J Pharm 2020; 589:119826. [PMID: 32871219 PMCID: PMC7836899 DOI: 10.1016/j.ijpharm.2020.119826] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/24/2020] [Accepted: 08/24/2020] [Indexed: 12/26/2022]
Abstract
Viral infections represent 44% of newly emerging infections, and as is shown by the COVID-19 outbreak constitute a major risk to human health and wellbeing. Although there are many efficient antiviral agents, they still have drawbacks such as development of virus resistance and accumulation within off-target organs. Encapsulation of antiviral agents into nanoparticles (NPs) has been shown to improve bioavailability, control release, and reduce side effects. However, there is little quantitative understanding of how the uptake of NPs into virally infected cells compares to uninfected cells. In this work, the uptake of fluorescently labeled polymer NPs was investigated in several models of rhinovirus (RV) infected cells. Different multiplicities of RV infections (MOI) and timings of NPs uptake were also investigated. In some cases, RV infection resulted in a significant increase of NPs uptake, but this was not universally noted. For HeLa cells, RV-A16 and RV-A01 infection elevated NPs uptake upon increasing the incubation time, whereas at later timepoints (6 h) a reduced uptake was noted with RV-A01 infection (owing to decreased cell viability). Beas-2B cells exhibited more complex trends: decreases in NPs uptake (cf. uninfected cells) were observed at short incubation times following RV-A01 and RV-A16 infection. At later incubation times (4 h), we found a marked decrease of NPs uptake for RV-A01 infected cells but an increase in uptake with RV-A16 infected cells. Where increases in NPs uptake were found, they were very modest compared to results previously reported for a hepatitis C/ Huh7.5 cell line model. An increase in RV dose (MOI) was not associated with any notable change of NPs uptake. We argue that the diverse endocytic pathways among the different cell lines, together with changes in virus nature, size, and entry mechanism are responsible for these differences. These findings suggest that NPs entry into virally infected cells is a complex process, and further work is required to unravel the different factors which govern this. Undertaking this additional research will be crucial to develop potent nanomedicines for the delivery of antiviral agents.
Collapse
Affiliation(s)
- Yasmin Abo-Zeid
- Department of Pharmaceutics, Faculty of Pharmacy, Helwan University, Cairo, Egypt; UCL School of Pharmacy, University College London, 29 - 39 Brunswick Square, London WC1N 1AX, UK; Cellular and Molecular Immunology Research Centre, London Metropolitan University, 166-220 Holloway Road, London N7 8DB, UK.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29 - 39 Brunswick Square, London WC1N 1AX, UK.
| | - Lila Touabi
- Cellular and Molecular Immunology Research Centre, London Metropolitan University, 166-220 Holloway Road, London N7 8DB, UK.
| | - Gary R McLean
- Cellular and Molecular Immunology Research Centre, London Metropolitan University, 166-220 Holloway Road, London N7 8DB, UK; National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, UK.
| |
Collapse
|
36
|
Khalili H, Nourian A, Ahmadinejad Z, Emadi Kouchak H, Jafari S, Dehghan Manshadi SA, Rasolinejad M, Kebriaeezadeh A. Efficacy and safety of sofosbuvir/ ledipasvir in treatment of patients with COVID-19; A randomized clinical trial. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:e2020102. [PMID: 33525212 PMCID: PMC7927527 DOI: 10.23750/abm.v91i4.10877] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
Background: There is no study regarding the use of SOF/LDP in treatment of COVID-19. Objectives: In this study, the efficacy and safety of SOF/LDP were assessed in treatment of patients with mild to moderate COVID-19. Methods: Among an open-label randomized clinical trial, 82 patients with mild to moderated COVID-19 were assigned to receive either SOF/LDP 400/100 mg daily plus the standard of care (SOF/LDP group, n=42) or the standard of care alone (control group, n=40) for 10 days. Time to clinical response, rate of clinical response, duration of hospital and ICU stay and 14-day mortality were assessed. Results: Clinical response occurred in 91.46% of patients. Although rates of clinical response were comparable between the groups but it occurred faster in the SOF/LDP group than the control group (2 vs. 4 days respectively, P= 0.02). Supportive cares were provided in the medical wards for most patients but 17.07% of patients were transferred to ICU during the hospitalization course. However, durations of hospital and ICU stay were comparable between the groups. 14-day mortality rate was 7.14% and 7.5% in the SOF/ LDP and control groups respectively. No adverse effects leading to drug discontinuation occurred. Gastrointestinal events (nausea, vomiting and diarrhea) were the most common side effects (15.85%). Conclusion: Added to the standard of care, SOF/LDP accelerated time to the clinical response. However, rate of clinical response, duration of hospital and ICU stay and 14-day mortality were not different. No significant adverse event was detected. More randomized clinical trials with larger sample sizes are needed to confirm the efficacy and safety of SOF/LDP in the treatment of COVID-19. (www.actabiomedica.it)
Collapse
Affiliation(s)
| | | | | | | | - Sirous Jafari
- Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | |
Collapse
|
37
|
Chien M, Anderson TK, Jockusch S, Tao C, Li X, Kumar S, Russo JJ, Kirchdoerfer RN, Ju J. Nucleotide Analogues as Inhibitors of SARS-CoV-2 Polymerase, a Key Drug Target for COVID-19. J Proteome Res 2020; 19:4690-4697. [PMID: 32692185 PMCID: PMC7640960 DOI: 10.1021/acs.jproteome.0c00392] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Indexed: 12/18/2022]
Abstract
SARS-CoV-2 is responsible for the current COVID-19 pandemic. On the basis of our analysis of hepatitis C virus and coronavirus replication, and the molecular structures and activities of viral inhibitors, we previously demonstrated that three nucleotide analogues (the triphosphates of Sofosbuvir, Alovudine, and AZT) inhibit the SARS-CoV RNA-dependent RNA polymerase (RdRp). We also demonstrated that a library of additional nucleotide analogues terminate RNA synthesis catalyzed by the SARS-CoV-2 RdRp, a well-established drug target for COVID-19. Here, we used polymerase extension experiments to demonstrate that the active triphosphate form of Sofosbuvir (an FDA-approved hepatitis C drug) is incorporated by SARS-CoV-2 RdRp and blocks further incorporation. Using the molecular insight gained from the previous studies, we selected the active triphosphate forms of six other antiviral agents, Alovudine, Tenofovir alafenamide, AZT, Abacavir, Lamivudine, and Emtricitabine, for evaluation as inhibitors of the SARS-CoV-2 RdRp and demonstrated the ability of these viral polymerase inhibitors to be incorporated by SARS-CoV-2 RdRp, where they terminate further polymerase extension with varying efficiency. These results provide a molecular basis for inhibition of the SARS-CoV-2 RdRp by these nucleotide analogues. If sufficient efficacy of some of these FDA-approved drugs in inhibiting viral replication in cell culture is established, they may be explored as potential COVID-19 therapeutics.
Collapse
Affiliation(s)
- Minchen Chien
- Center
for Genome Technology and Biomolecular Engineering, Departments of Chemical
Engineering, Pharmacology, and Chemistry, Columbia University, New York, New York 10027, United States
| | - Thomas K. Anderson
- Department of Biochemistry and Institute of Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Steffen Jockusch
- Center
for Genome Technology and Biomolecular Engineering, Departments of Chemical
Engineering, Pharmacology, and Chemistry, Columbia University, New York, New York 10027, United States
| | - Chuanjuan Tao
- Center
for Genome Technology and Biomolecular Engineering, Departments of Chemical
Engineering, Pharmacology, and Chemistry, Columbia University, New York, New York 10027, United States
| | - Xiaoxu Li
- Center
for Genome Technology and Biomolecular Engineering, Departments of Chemical
Engineering, Pharmacology, and Chemistry, Columbia University, New York, New York 10027, United States
| | - Shiv Kumar
- Center
for Genome Technology and Biomolecular Engineering, Departments of Chemical
Engineering, Pharmacology, and Chemistry, Columbia University, New York, New York 10027, United States
| | - James J. Russo
- Center
for Genome Technology and Biomolecular Engineering, Departments of Chemical
Engineering, Pharmacology, and Chemistry, Columbia University, New York, New York 10027, United States
| | - Robert N. Kirchdoerfer
- Department of Biochemistry and Institute of Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Jingyue Ju
- Center
for Genome Technology and Biomolecular Engineering, Departments of Chemical
Engineering, Pharmacology, and Chemistry, Columbia University, New York, New York 10027, United States
| |
Collapse
|
38
|
Jockusch S, Tao C, Li X, Chien M, Kumar S, Morozova I, Kalachikov S, Russo JJ, Ju J. Sofosbuvir terminated RNA is more resistant to SARS-CoV-2 proofreader than RNA terminated by Remdesivir. Sci Rep 2020; 10:16577. [PMID: 33024223 PMCID: PMC7538426 DOI: 10.1038/s41598-020-73641-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022] Open
Abstract
SARS-CoV-2 is responsible for COVID-19, resulting in the largest pandemic in over a hundred years. After examining the molecular structures and activities of hepatitis C viral inhibitors and comparing hepatitis C virus and coronavirus replication, we previously postulated that the FDA-approved hepatitis C drug EPCLUSA (Sofosbuvir/Velpatasvir) might inhibit SARS-CoV-2. We subsequently demonstrated that Sofosbuvir triphosphate is incorporated by the relatively low fidelity SARS-CoV and SARS-CoV-2 RNA-dependent RNA polymerases (RdRps), serving as an immediate polymerase reaction terminator, but not by a host-like high fidelity DNA polymerase. Other investigators have since demonstrated the ability of Sofosbuvir to inhibit SARS-CoV-2 replication in lung and brain cells; additionally, COVID-19 clinical trials with EPCLUSA and with Sofosbuvir plus Daclatasvir have been initiated in several countries. SARS-CoV-2 has an exonuclease-based proofreader to maintain the viral genome integrity. Any effective antiviral targeting the SARS-CoV-2 RdRp must display a certain level of resistance to this proofreading activity. We report here that Sofosbuvir terminated RNA resists removal by the exonuclease to a substantially higher extent than RNA terminated by Remdesivir, another drug being used as a COVID-19 therapeutic. These results offer a molecular basis supporting the current use of Sofosbuvir in combination with other drugs in COVID-19 clinical trials.
Collapse
Affiliation(s)
- Steffen Jockusch
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Chuanjuan Tao
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA
- Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Xiaoxu Li
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA
- Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Minchen Chien
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA
- Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Shiv Kumar
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA
- Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Irina Morozova
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA
- Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Sergey Kalachikov
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA
- Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - James J Russo
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA
- Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Jingyue Ju
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA.
- Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA.
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
39
|
Kardani K, Basimi P, Fekri M, Bolhassani A. Antiviral therapy for the sexually transmitted viruses: recent updates on vaccine development. Expert Rev Clin Pharmacol 2020; 13:1001-1046. [PMID: 32838584 DOI: 10.1080/17512433.2020.1814743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The sexually transmitted infections (STIs) caused by viruses including human T cell leukemia virus type-1 (HTLV-1), human immunodeficiency virus-1 (HIV-1), human simplex virus-2 (HSV-2), hepatitis C virus (HCV), hepatitis B virus (HBV), and human papillomavirus (HPV) are major public health issues. These infections can cause cancer or result in long-term health problems. Due to high prevalence of STIs, a safe and effective vaccine is required to overcome these fatal viruses. AREAS COVERED This review includes a comprehensive overview of the literatures relevant to vaccine development against the sexually transmitted viruses (STVs) using PubMed and Sciencedirect electronic search engines. Herein, we discuss the efforts directed toward development of effective vaccines using different laboratory animal models including mice, guinea pig or non-human primates in preclinical trials, and human in clinical trials with different phases. EXPERT OPINION There is no effective FDA approved vaccine against the sexually transmitted viruses (STVs) except for HBV and HPV as prophylactic vaccines. Many attempts are underway to develop vaccines against these viruses. There are several approaches for improving prophylactic or therapeutic vaccines such as heterologous prime/boost immunization, delivery system, administration route, adjuvants, etc. In this line, further studies can be helpful for understanding the immunobiology of STVs in human. Moreover, development of more relevant animal models is a worthy goal to induce effective immune responses in humans.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| | - Parya Basimi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| | - Mehrshad Fekri
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| |
Collapse
|
40
|
Jockusch S, Tao C, Li X, Anderson TK, Chien M, Kumar S, Russo JJ, Kirchdoerfer RN, Ju J. A library of nucleotide analogues terminate RNA synthesis catalyzed by polymerases of coronaviruses that cause SARS and COVID-19. Antiviral Res 2020; 180:104857. [PMID: 32562705 PMCID: PMC7299870 DOI: 10.1016/j.antiviral.2020.104857] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/30/2020] [Accepted: 06/12/2020] [Indexed: 02/08/2023]
Abstract
SARS-CoV-2, a member of the coronavirus family, is responsible for the current COVID-19 worldwide pandemic. We previously demonstrated that five nucleotide analogues inhibit the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp), including the active triphosphate forms of Sofosbuvir, Alovudine, Zidovudine, Tenofovir alafenamide and Emtricitabine. We report here the evaluation of a library of nucleoside triphosphate analogues with a variety of structural and chemical features as inhibitors of the RdRps of SARS-CoV and SARS-CoV-2. These features include modifications on the sugar (2' or 3' modifications, carbocyclic, acyclic, or dideoxynucleotides) or on the base. The goal is to identify nucleotide analogues that not only terminate RNA synthesis catalyzed by these coronavirus RdRps, but also have the potential to resist the viruses' exonuclease activity. We examined these nucleotide analogues for their ability to be incorporated by the RdRps in the polymerase reaction and to prevent further incorporation. While all 11 molecules tested displayed incorporation, 6 exhibited immediate termination of the polymerase reaction (triphosphates of Carbovir, Ganciclovir, Stavudine and Entecavir; 3'-OMe-UTP and Biotin-16-dUTP), 2 showed delayed termination (Cidofovir diphosphate and 2'-OMe-UTP), and 3 did not terminate the polymerase reaction (2'-F-dUTP, 2'-NH2-dUTP and Desthiobiotin-16-UTP). The coronaviruses possess an exonuclease that apparently requires a 2'-OH at the 3'-terminus of the growing RNA strand for proofreading. In this study, all nucleoside triphosphate analogues evaluated form Watson-Crick-like base pairs. The nucleotide analogues demonstrating termination either lack a 2'-OH, have a blocked 2'-OH, or show delayed termination. Thus, these nucleotide analogues are of interest for further investigation to evaluate whether they can evade the viral exonuclease activity. Prodrugs of five of these nucleotide analogues (Cidofovir, Abacavir, Valganciclovir/Ganciclovir, Stavudine and Entecavir) are FDA-approved medications for treatment of other viral infections, and their safety profiles are well established. After demonstrating potency in inhibiting viral replication in cell culture, candidate molecules can be rapidly evaluated as potential therapies for COVID-19.
Collapse
Affiliation(s)
- Steffen Jockusch
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Chuanjuan Tao
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Xiaoxu Li
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Thomas K Anderson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Institute of Molecular Virology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Minchen Chien
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Shiv Kumar
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - James J Russo
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Robert N Kirchdoerfer
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Institute of Molecular Virology, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Jingyue Ju
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA; Department of Pharmacology, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
41
|
Sun CF, Chiu WC, Chen PC, Chang HC, Guu TW, Shityakov S, Miller AH, Felger JC, Chang JPC, Su KP. Depression-free after Interferon-α exposure indicates less incidence of depressive disorder: A longitudinal study in Taiwan. Brain Behav Immun 2020; 88:125-131. [PMID: 32434045 DOI: 10.1016/j.bbi.2020.05.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 10/24/2022] Open
Abstract
BACKGROUND IFN-α-induced depression in patients undergoing hepatitis C virus (HCV) treatment provides powerful support for the inflammation hypothesis of depression. Most studies have focused on the occurrence of depressive symptoms, but there has been no study yet in depression-free HCV patients receiving IFN-α. We hypothesized that HCV patients who did not develop depression after IFN-α exposure might have a lower incidence of depressive disorders after the IFN-α treatment. METHODS We conducted a twelve-year population-based cohort study of chronic HCV patients who received IFN-α therapy. The data were obtained from the Taiwan National Health Insurance Research Database. The study cohort was patients without any depressive disorder nor antidepressant use before and during IFN-α therapy. They were matched randomly by age, sex income and urbanization at a ratio of 1:4 with the control cohort of HCV patients without IFN-α therapy. The follow-up started after the last administration of IFN-α, and the primary outcome was the incidence of depressive disorders after IFN-α therapy. RESULTS A total of 20,468 depression-free subjects were identified from records of HCV patients receiving IFN-α therapy. Patients without IFN-α-induced depression were associated with a significantly lower incidence (per 10,000 person-years) of new-onset depressive disorders (126.8, 95% Confidential Interval [CI] of 118.5-135.6) as compared to the control cohort (145.2, 95% CI of 140.0-150.6) (p < 0.001). After adjusting for age, sex, income, urbanization and comorbid diseases, the crude hazard ratio for the incident depressive disorder was 0.87 (95% CI, 0.80-0.87) and the adjusted hazard ratios was 0.79 (95% CI, 0.72-0.87) for IFN-α-induced depression-free subjects as compared to the controls. DISCUSSION Our study indicates that IFN-α treated depression-free patients have a lower risk for depressive disorders. This hypothesized mechanism might derive from an IFN-α-induced resilience factor as yet to be defined. CONCLUSIONS Our study might suggest a new possibility for a new pharmacological strategy against depression.
Collapse
Affiliation(s)
- Ching-Fang Sun
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Che Chiu
- Department of Psychiatry, Cathay General Hospital, Taipei, Taiwan; School of Medicine, Fu Jen Catholic University, Taipei, Taiwan.
| | - Pau-Chung Chen
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health, Taipei, Taiwan; Department of Public Health, National Taiwan University College of Public Health, Taipei, Taiwan; Department of Environmental and Occupational Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hui-Chih Chang
- School of Health Care Administration, Taipei Medical University, Taiwan; Department of Psychological Medicine, Institute of Psychiatry, King's College London, UK
| | - Ta-Wei Guu
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Division of Psychiatry, Departments of Internal Medicine, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Sergey Shityakov
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Department of Bioinformatics, University of Würzburg, 97080 Würzburg, Germany
| | - Andrew H Miller
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Jane Pei-Chen Chang
- College of Medicine, China Medical University, Taichung, Taiwan; Department of Psychological Medicine, Institute of Psychiatry, King's College London, UK; Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
| | - Kuan-Pin Su
- College of Medicine, China Medical University, Taichung, Taiwan; Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; An-Nan Hospital, China Medical University, Tainan, Taiwan
| |
Collapse
|
42
|
Hopkins L, Dunlap T, Cline H. Pharmacology Update for the Treatment of Hepatitis C Virus. Nurs Clin North Am 2020; 55:347-359. [PMID: 32762855 DOI: 10.1016/j.cnur.2020.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) is a common infectious disease affecting people worldwide. In the past 10 years, the incidence of HCV has steadily increased in the United States. With the advent of new direct-acting antiviral medications, the treatment of HCV has become important and can cure the infection.
Collapse
Affiliation(s)
- Leslie Hopkins
- Vanderbilt University School of Nursing, 461 21st Avenue South, Nashville, TN 37240, USA.
| | - Travis Dunlap
- Vanderbilt University School of Nursing, 461 21st Avenue South, Nashville, TN 37240, USA
| | - Holly Cline
- Vanderbilt University School of Nursing, 461 21st Avenue South, Nashville, TN 37240, USA
| |
Collapse
|
43
|
Lim SK, Othman R, Yusof R, Heh CH. Rational drug discovery: Ellagic acid as a potent dual-target inhibitor against hepatitis C virus genotype 3 (HCV G3) NS3 enzymes. Chem Biol Drug Des 2020; 97:28-40. [PMID: 32657543 DOI: 10.1111/cbdd.13756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 06/15/2020] [Accepted: 06/28/2020] [Indexed: 12/19/2022]
Abstract
Structure-based virtual screening (SBVS) has served as a popular strategy for rational drug discovery. In this study, we aimed to discover novel benzopyran-based inhibitors that targeted the NS3 enzymes (NS3/4A protease and NS3 helicase) of HCV G3 using a combination of in silico and in vitro approaches. With the aid of SBVS, six novel compounds were discovered to inhibit HCV G3 NS3/4A protease and two phytochemicals (ellagic acid and myricetin) were identified as dual-target inhibitors that inhibited both NS3/4A protease and NS3 helicase in vitro (IC50 = 40.37 ± 5.47 nm and 6.58 ± 0.99 µm, respectively). Inhibitory activities against the replication of HCV G3 replicons were further assessed in a cell-based system with four compounds showed dose-dependent inhibition. Compound P8 was determined to be the most potent compound from the cell-based assay with an EC50 of 19.05 µm. The dual-target inhibitor, ellagic acid, was determined as the second most potent (EC50 = 32.37 µm) and the most selective in its inhibitory activity against the replication of HCV replicons, without severely affecting the viability of the host cells (selectivity index > 6.18).
Collapse
Affiliation(s)
- See Khai Lim
- Drug Design and Development Research Group, University of Malaya, Kuala Lumpur, Malaysia.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Rozana Othman
- Drug Design and Development Research Group, University of Malaya, Kuala Lumpur, Malaysia.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia.,Center for Natural Products Research and Drug Discovery (CENAR), University of Malaya, Kuala Lumpur, Malaysia
| | - Rohana Yusof
- Drug Design and Development Research Group, University of Malaya, Kuala Lumpur, Malaysia.,Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Choon Han Heh
- Drug Design and Development Research Group, University of Malaya, Kuala Lumpur, Malaysia.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
44
|
Abstract
Hepatitis C virus (HCV) accounts for hepatitis, cirrhosis, hepatocellular carcinoma, and liver transplantation. Every year, millions of people develop chronic liver diseases. This article provides novel insights into the major breakthroughs in the discovery of hepatitis C virus. 2020 Nobel Prize in Medicine was awarded to Michael Houghton, Harvey Alter, and Charles Rice for their outstanding contribution in the discovery of HCV. Also, this article deals with current findings, challenges, and future prospects in the diagnosis of HCV infection. DAA, along with protease inhibitors, is found to have higher SVR and is a promising drug to treat HCV infection. However, with regard to the emergence of resistance to DAAs, CRISPR-Cas can be the future technique in preventing resistance. Although the current knowledge of molecular mechanisms associated with HCV infection is insufficient, the better understanding of it provides better hope in the treatment of HCV. Lymphoma A cancer causing infection on lymphocytes. Fulminant hepatitis A decrease in the size of the liver accompanied even by the death of liver parenchyma with the onset of infection with hepatitis virus. Transfection The process of introducing nucleic acids into eukaryotic cells without using viral methods. Huh-7 cells A type of liver cells grown in the laboratory for research purposes. Immunoscreening A biomedical method that helps to detect a protein produced from a cloned gene after it undergoes transcription, followed by the translation process within the cell. cDNA library A group of only the genes that are encoded into proteins by an organism. Sustained virologic response Absence of any evidence of the presence of HCV in the blood of patients with chronic HCV infection after undergoing antiviral treatment. Pegylated interferon A covalent conjugate of recombinant interferon and polyethylene glycol, used as an antiviral and antineoplastic agent. Immunoelectron microscopy A technique to localize ultrastructure antigens or antibodies in cells or tissues for the diagnosis of viral infections. Stellate cells A major type of liver cell involved in liver cirrhosis. Hepatitis C virus (HCV) accounts for hepatitis, liver cirrhosis, hepatocellular carcinoma, and liver transplantation. This virus is a single-stranded RNA virus that belongs to the Flaviviridae family. According to the WHO, about 71 million people have chronic HCV infections around the globe in 2020, and hence, it is a plague of humankind. The credit of discovery of HCV goes to Michael Houghton, Harvey Alter, and Charles Rice for which they are awarded 2020 Nobel Prize in Medicine. Their contribution has given better hope to mankind to cure HCV for the first time in the history. With the use of pegylated interferon and ribavirin jointly, higher SVR has been found comparatively, even in patients with chronic liver diseases. However, due to excessive pain tolerated by patients, interferon (IFN)-based therapy is rapidly being replaced with IFN-free DAA regimens. With the onset of resistance to DAA drugs, CRISPR-Cas system can be used to modify the viral genome to impair their ability to develop resistance. How to cite this article: Laugi H. Discovery of Hepatitis C Virus: 2020 Nobel Prize in Medicine. Euroasian J Hepato-Gastroenterol 2020;10(2): 105–108.
Collapse
|
45
|
Martínez JL, Arias CF. Role of the Guanine Nucleotide Exchange Factor GBF1 in the Replication of RNA Viruses. Viruses 2020; 12:E682. [PMID: 32599855 PMCID: PMC7354614 DOI: 10.3390/v12060682] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
The guanine nucleotide exchange factor GBF1 is a well-known factor that can activate different ADP-ribosylation factor (Arf) proteins during the regulation of different cellular vesicular transport processes. In the last decade, it has become increasingly evident that GBF1 can also regulate different steps of the replication cycle of RNA viruses belonging to different virus families. GBF1 has been shown not only to facilitate the intracellular traffic of different viral and cellular elements during infection, but also to modulate the replication of viral RNA, the formation and maturation of viral replication complexes, and the processing of viral proteins through mechanisms that do not depend on its canonical role in intracellular transport. Here, we review the various roles that GBF1 plays during the replication of different RNA viruses.
Collapse
Affiliation(s)
| | - Carlos F. Arias
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 4510, Morelos, Mexico;
| |
Collapse
|
46
|
|
47
|
Mohammadzadeh I, Qujeq D, Yousefi T, Ferns GA, Maniati M, Vaghari-Tabari M. CRISPR/Cas9 gene editing: A new therapeutic approach in the treatment of infection and autoimmunity. IUBMB Life 2020; 72:1603-1621. [PMID: 32344465 DOI: 10.1002/iub.2296] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022]
Abstract
CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein9) may be viewed as an adaptive bacterial immune system. When a virus infects a bacterium, a fragment of the virus genome is inserted into the CRISPR sequence of the bacterial genome as a memory. When the bacterium becomes infected again with the same virus, an RNA molecule that is a transcript of the memory sequence, directs Cas9, an endonuclease, to the complementary region of the virus genome, and Cas9 disables the virus by a double-strand break. In recent years, studies have shown that by designing synthetic RNA molecules and delivering them along with Cas9 into eukaryotic cells, different regions of the cell's genome can be targeted and manipulated. These findings have drawn much attention to this new technology and it has been shown that CRISPR/Cas9 gene editing can be used to treat some human diseases. These include infectious diseases and autoimmune diseases. In this review article, in addition to a brief overview of the biology of the CRISPR/Cas9 system, we collected the most recent findings on the applications of CRISPR/Cas9 technology for better investigation of the pathogenesis and treatment of viral infections (human immunodeficiency virus infection, hepatitis virus infections, and onco-virus infections), non-viral infections (parasitic, fungal, and bacterial infections), and autoimmune diseases.
Collapse
Affiliation(s)
- Iraj Mohammadzadeh
- Non-Communicable Pediatric Diseases Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Tooba Yousefi
- Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Gordon A Ferns
- Department of Medical Education, Brighton & Sussex Medical School, Brighton, UK
| | - Mahmood Maniati
- English Department, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
48
|
Chien M, Anderson TK, Jockusch S, Tao C, Kumar S, Li X, Russo JJ, Kirchdoerfer RN, Ju J. Nucleotide Analogues as Inhibitors of SARS-CoV-2 Polymerase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.03.18.997585. [PMID: 32511320 PMCID: PMC7239050 DOI: 10.1101/2020.03.18.997585] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SARS-CoV-2, a member of the coronavirus family, is responsible for the current COVID-19 pandemic. Based on our analysis of hepatitis C virus and coronavirus replication, and the molecular structures and activities of viral inhibitors, we previously demonstrated that three nucleotide analogues inhibit the SARS-CoV RNA-dependent RNA polymerase (RdRp). Here, using polymerase extension experiments, we have demonstrated that the active triphosphate form of Sofosbuvir (a key component of the FDA approved hepatitis C drug EPCLUSA), is incorporated by SARS-CoV-2 RdRp, and blocks further incorporation. Using the same molecular insight, we selected the active triphosphate forms of three other anti-viral agents, Alovudine, AZT (an FDA approved HIV/AIDS drug) and Tenofovir alafenamide (TAF, an FDA approved drug for HIV and hepatitis B) for evaluation as inhibitors of SARS-CoV-2 RdRp. We demonstrated the ability of these three viral polymerase inhibitors, 3'-fluoro-3'-deoxythymidine triphosphate, 3'-azido-3'-deoxythymidine triphosphate and Tenofovir diphosphate (the active triphosphate forms of Alovudine, AZT and TAF, respectively) to be incorporated by SARS-CoV-2 RdRp, where they also terminate further polymerase extension. These results offer a strong molecular basis for these nucleotide analogues to be evaluated as potential therapeutics for COVID-19.
Collapse
Affiliation(s)
- Minchen Chien
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY 10027
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | - Thomas K. Anderson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
- Institute of Molecular Virology, University of Wisconsin-Madison, Madison, WI 53706
| | - Steffen Jockusch
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY 10027
- Department of Chemistry, Columbia University, New York, NY 10027
| | - Chuanjuan Tao
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY 10027
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | - Shiv Kumar
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY 10027
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | - Xiaoxu Li
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY 10027
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | - James J. Russo
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY 10027
- Department of Chemical Engineering, Columbia University, New York, NY 10027
| | - Robert N. Kirchdoerfer
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
- Institute of Molecular Virology, University of Wisconsin-Madison, Madison, WI 53706
| | - Jingyue Ju
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY 10027
- Department of Chemical Engineering, Columbia University, New York, NY 10027
- Department of Pharmacology, Columbia University, New York, NY 10027
| |
Collapse
|
49
|
|
50
|
Managing sexually transmitted infections: Beyond the 2015 guidelines. Nurse Pract 2019; 43:28-34. [PMID: 29985197 DOI: 10.1097/01.npr.0000541464.23795.5b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Guidelines for the prevention and management of sexually transmitted infections (STIs) are updated periodically while new science is continuously developed. Advanced practice registered nurses implement clinical decisions based on current guidelines and evidence. This article provides recent updates on managing STIs.
Collapse
|