1
|
Ying Q, Zhang X, Gu T, Zhang J, Dong Y, Feng W, Li D, Wu X, Wang F. Apatinib inhibits HTNV by stimulating TFEB-driven lysosome biogenesis to degrade viral protein. Antiviral Res 2025; 237:106124. [PMID: 40020878 DOI: 10.1016/j.antiviral.2025.106124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Hantaan Orthohantavirus (Hantaan virus, HTNV) infection causes hemorrhagic fever with renal syndrome (HFRS) in humans, posing a significant health threat. Currently, there are no long-lasting protective vaccines or specific antivirals available, creating an urgent need for effective antiviral treatments in the clinical management of HFRS. Given that viruses exploit multiple host factors for their replication, host-oriented inhibitors could offer promising therapeutic options. In our study, we screened a library of 2570 drugs and identified apatinib, a kinase inhibitor, as a potent suppressor of HTNV infection both in vitro and in vivo. Mechanistic studies revealed that apatinib exerts its antiviral effect by targeting transcription factor EB (TFEB). Specifically, apatinib inhibits the PI3K-Akt signaling pathway and reduces mTOR phosphorylation, which in turn downregulates TFEB phosphorylation. This facilitates the nuclear translocation of TFEB and enhances lysosomal function by upregulating the expression of lysosome-associated genes and promoting lysosome biogenesis. Consequently, there is an increase in lysosome-mediated viral nucleocapsid protein degradation. The ability of apatinib to stimulate this lysosome-driven antiviral mechanism presents a potential new therapeutic approach for viral infections and offers valuable insights into virus-host interactions during HTNV replication.
Collapse
Affiliation(s)
- Qikang Ying
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Xiaoxiao Zhang
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Tianle Gu
- Department of Pathogen Biology, College of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Junmei Zhang
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Yuhang Dong
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Wenjie Feng
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Dongjing Li
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Xingan Wu
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China.
| | - Fang Wang
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China.
| |
Collapse
|
2
|
Bronowicka-Szydełko A, Rabczyński M, Dumas I, Fiodorenko-Dumas Ż, Wojtczak B, Kotyra Ł, Kustrzeba-Wójcicka I, Lewandowski Ł, Ponikowska B, Kuzan A, Kluz J, Gamian A, Madziarska K. State of Knowledge About Thyroid Cancers in the Era of COVID-19-A Narrative Review. Biomedicines 2024; 12:2829. [PMID: 39767735 PMCID: PMC11672969 DOI: 10.3390/biomedicines12122829] [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: 10/26/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 01/03/2025] Open
Abstract
Thyroid cancer (TC), due to its heterogeneous nature, remains a clinical challenge. Many factors can initiate the carcinogenesis process of various types of TC, which complicates diagnosis and treatment. The presented review gathers current information on specific types of TC, taking into account the effects of the COVID-19 pandemic. It is likely that COVID-19 has influenced and continues to influence the function of the thyroid gland. A high percentage of patients with COVID-19 showing simultaneous pathological changes in the thyroid suggests that SARS-CoV-2 may disrupt the function of this gland and initiate pro-oxidative mechanisms, inflammatory states, and autoimmune diseases, thereby promoting the formation of neoplastic changes. Furthermore, changes in the expression of the ACE2, TMPRSS2, CLEC4M and DPP4 genes, observed in TC, also occur in COVID-19. Therefore, it is probable that the interaction of SARS-CoV-2 with thyroid cell receptors may initiate carcinogenesis in this gland. Additionally, some drugs used in TC therapy (e.g., levothyroxine) may increase the affinity of SARS-CoV-2 for cells, which could contribute to a more severe course of COVID-19 and the emergence of long-term symptoms (post-COVID-19). Moreover, the consequences of sanitary restrictions (limited access to medical services, reduction in endocrinological and oncological procedures) that took place in many countries during the COVID-19 pandemic may lead in the future to an increased number of missed diagnoses and the emergence of aggressive cancers.
Collapse
Affiliation(s)
- Agnieszka Bronowicka-Szydełko
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.B.-S.); (Ł.K.); (I.K.-W.); (Ł.L.)
| | - Maciej Rabczyński
- Clinical Department of Diabetology, Hypertension and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (M.R.); (J.K.); (K.M.)
| | - Ilias Dumas
- Department of Clinical Physiotherapy and Rehabilitation, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Żanna Fiodorenko-Dumas
- Department of Clinical Physiotherapy and Rehabilitation, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Beata Wojtczak
- University Center for General and Oncological Surgery, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Łukasz Kotyra
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.B.-S.); (Ł.K.); (I.K.-W.); (Ł.L.)
| | - Irena Kustrzeba-Wójcicka
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.B.-S.); (Ł.K.); (I.K.-W.); (Ł.L.)
| | - Łukasz Lewandowski
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.B.-S.); (Ł.K.); (I.K.-W.); (Ł.L.)
| | - Beata Ponikowska
- Department of Physiology and Pathophysiology, Division of Physiology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Aleksandra Kuzan
- Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Wroclaw University of Science and Technology, 51-377 Wroclaw, Poland;
| | - Joanna Kluz
- Clinical Department of Diabetology, Hypertension and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (M.R.); (J.K.); (K.M.)
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimantal Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Katarzyna Madziarska
- Clinical Department of Diabetology, Hypertension and Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (M.R.); (J.K.); (K.M.)
| |
Collapse
|
3
|
Liao J, Yi H, Wang H, Yang S, Jiang D, Huang X, Zhang M, Shen J, Lu H, Niu Y. CDCM: a correlation-dependent connectivity map approach to rapidly screen drugs during outbreaks of infectious diseases. Brief Bioinform 2024; 26:bbae659. [PMID: 39701599 DOI: 10.1093/bib/bbae659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/06/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
In the context of the global damage caused by coronavirus disease 2019 (COVID-19) and the emergence of the monkeypox virus (MPXV) outbreak as a public health emergency of international concern, research into methods that can rapidly test potential therapeutics during an outbreak of a new infectious disease is urgently needed. Computational drug discovery is an effective way to solve such problems. The existence of various large open databases has mitigated the time and resource consumption of traditional drug development and improved the speed of drug discovery. However, the diversity of cell lines used in various databases remains limited, and previous drug discovery methods are ineffective for cross-cell prediction. In this study, we propose a correlation-dependent connectivity map (CDCM) to achieve cross-cell predictions of drug similarity. The CDCM mainly identifies drug-drug or disease-drug relationships from the perspective of gene networks by exploring the correlation changes between genes and identifying similarities in the effects of drugs or diseases on gene expression. We validated the CDCM on multiple datasets and found that it performed well for drug identification across cell lines. A comparison with the Connectivity Map revealed that our method was more stable and performed better across different cell lines. In the application of the CDCM to COVID-19 and MPXV data, the predictions of potential therapeutic compounds for COVID-19 were consistent with several previous studies, and most of the predicted drugs were found to be experimentally effective against MPXV. This result confirms the practical value of the CDCM. With the ability to predict across cell lines, the CDCM outperforms the Connectivity Map, and it has wider application prospects and a reduced cost of use.
Collapse
Affiliation(s)
- Junlei Liao
- School of Mathematics and Statistics, HNP-LAMA, Central South University, Changsha 410083, Hunan, China
| | - Hongyang Yi
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Hao Wang
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen 518133, China
| | - Sumei Yang
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Duanmei Jiang
- School of Mathematics and Statistics, HNP-LAMA, Central South University, Changsha 410083, Hunan, China
| | - Xin Huang
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen 518133, China
| | - Mingxia Zhang
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Jiayin Shen
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Hongzhou Lu
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, China
| | - Yuanling Niu
- School of Mathematics and Statistics, HNP-LAMA, Central South University, Changsha 410083, Hunan, China
| |
Collapse
|
4
|
Freidel MR, Vakhariya PA, Sardarni SK, Armen RS. The Dual-Targeted Fusion Inhibitor Clofazimine Binds to the S2 Segment of the SARS-CoV-2 Spike Protein. Viruses 2024; 16:640. [PMID: 38675980 PMCID: PMC11054727 DOI: 10.3390/v16040640] [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: 03/01/2024] [Revised: 03/29/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Clofazimine and Arbidol have both been reported to be effective in vitro SARS-CoV-2 fusion inhibitors. Both are promising drugs that have been repurposed for the treatment of COVID-19 and have been used in several previous and ongoing clinical trials. Small-molecule bindings to expressed constructs of the trimeric S2 segment of Spike and the full-length SARS-CoV-2 Spike protein were measured using a Surface Plasmon Resonance (SPR) binding assay. We demonstrate that Clofazimine, Toremifene, Arbidol and its derivatives bind to the S2 segment of the Spike protein. Clofazimine provided the most reliable and highest-quality SPR data for binding with S2 over the conditions explored. A molecular docking approach was used to identify the most favorable binding sites on the S2 segment in the prefusion conformation, highlighting two possible small-molecule binding sites for fusion inhibitors. Results related to molecular docking and modeling of the structure-activity relationship (SAR) of a newly reported series of Clofazimine derivatives support the proposed Clofazimine binding site on the S2 segment. When the proposed Clofazimine binding site is superimposed with other experimentally determined coronavirus structures in structure-sequence alignments, the changes in sequence and structure may rationalize the broad-spectrum antiviral activity of Clofazimine in closely related coronaviruses such as SARS-CoV, MERS, hCoV-229E, and hCoV-OC43.
Collapse
Affiliation(s)
| | | | | | - Roger S. Armen
- Department of Pharmaceutical Sciences, College of Pharmacy, Thomas Jefferson University, 901 Walnut St. Suite 918, Philadelphia, PA 19170, USA (P.A.V.); (S.K.S.)
| |
Collapse
|
5
|
Witwit H, Khafaji R, Salaniwal A, Kim AS, Cubitt B, Jackson N, Ye C, Weiss SR, Martinez-Sobrido L, de la Torre JC. Activation of protein kinase receptor (PKR) plays a pro-viral role in mammarenavirus-infected cells. J Virol 2024; 98:e0188323. [PMID: 38376197 PMCID: PMC10949842 DOI: 10.1128/jvi.01883-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/26/2024] [Indexed: 02/21/2024] Open
Abstract
Many viruses, including mammarenaviruses, have evolved mechanisms to counteract different components of the host cell innate immunity, which is required to facilitate robust virus multiplication. The double-stranded RNA (dsRNA) sensor protein kinase receptor (PKR) pathway plays a critical role in the cell anti-viral response. Whether PKR can restrict the multiplication of the Old World mammarenavirus lymphocytic choriomeningitis virus (LCMV) and the mechanisms by which LCMV may counteract the anti-viral functions of PKR have not yet been investigated. Here we present evidence that LCMV infection results in very limited levels of PKR activation, but LCMV multiplication is enhanced in the absence of PKR. In contrast, infection with a recombinant LCMV with a mutation affecting the 3'-5' exonuclease (ExoN) activity of the viral nucleoprotein resulted in robust PKR activation in the absence of detectable levels of dsRNA, which was associated with severely restricted virus multiplication that was alleviated in the absence of PKR. However, pharmacological inhibition of PKR activation resulted in reduced levels of LCMV multiplication. These findings uncovered a complex role of the PKR pathway in LCMV-infected cells involving both pro- and anti-viral activities.IMPORTANCEAs with many other viruses, the prototypic Old World mammarenavirus LCMV can interfere with the host cell innate immune response to infection, which includes the dsRNA sensor PKR pathway. A detailed understanding of LCMV-PKR interactions can provide novel insights about mammarenavirus-host cell interactions and facilitate the development of effective anti-viral strategies against human pathogenic mammarenaviruses. In the present work, we present evidence that LCMV multiplication is enhanced in PKR-deficient cells, but pharmacological inhibition of PKR activation unexpectedly resulted in severely restricted propagation of LCMV. Likewise, we document a robust PKR activation in LCMV-infected cells in the absence of detectable levels of dsRNA. Our findings have revealed a complex role of the PKR pathway during LCMV infection and uncovered the activation of PKR as a druggable target for the development of anti-viral drugs against human pathogenic mammarenaviruses.
Collapse
Affiliation(s)
- Haydar Witwit
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Roaa Khafaji
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Arul Salaniwal
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Arthur S. Kim
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, USA
| | - Beatrice Cubitt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | | | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Susan R. Weiss
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
6
|
Utrilla-Trigo S, Jiménez-Cabello L, Marín-López A, Illescas-Amo M, Andrés G, Calvo-Pinilla E, Lorenzo G, van Rijn PA, Ortego J, Nogales A. Engineering recombinant replication-competent bluetongue viruses expressing reporter genes for in vitro and non-invasive in vivo studies. Microbiol Spectr 2024; 12:e0249323. [PMID: 38353566 PMCID: PMC10923215 DOI: 10.1128/spectrum.02493-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 12/22/2023] [Indexed: 03/06/2024] Open
Abstract
Bluetongue virus (BTV) is the causative agent of the important livestock disease bluetongue (BT), which is transmitted via Culicoides bites. BT causes severe economic losses associated with its considerable impact on health and trade of animals. By reverse genetics, we have designed and rescued reporter-expressing recombinant (r)BTV expressing NanoLuc luciferase (NLuc) or Venus fluorescent protein. To generate these viruses, we custom synthesized a modified viral segment 5 encoding NS1 protein with the reporter genes located downstream and linked by the Porcine teschovirus-1 (PTV-1) 2A autoproteolytic cleavage site. Therefore, fluorescent signal or luciferase activity is only detected after virus replication and expression of non-structural proteins. Fluorescence or luminescence signals were detected in cells infected with rBTV/Venus or rBTV/NLuc, respectively. Moreover, the marking of NS2 protein confirmed that reporter genes were only expressed in BTV-infected cells. Growth kinetics of rBTV/NLuc and rBTV/Venus in Vero cells showed replication rates similar to those of wild-type and rBTV. Infectivity studies of these recombinant viruses in IFNAR(-/-) mice showed a higher lethal dose for rBTV/NLuc and rBTV/Venus than for rBTV indicating that viruses expressing the reporter genes are attenuated in vivo. Interestingly, luciferase activity was detected in the plasma of viraemic mice infected with rBTV/NLuc. Furthermore, luciferase activity quantitatively correlated with RNAemia levels of infected mice throughout the infection. In addition, we have investigated the in vivo replication and dissemination of BTV in IFNAR (-/-) mice using BTV/NLuc and non-invasive in vivo imaging systems.IMPORTANCEThe use of replication-competent viruses that encode a traceable fluorescent or luciferase reporter protein has significantly contributed to the in vitro and in vivo study of viral infections and the development of novel therapeutic approaches. In this work, we have generated rBTV that express fluorescent or luminescence proteins to track BTV infection both in vitro and in vivo. Despite the availability of vaccines, BTV and other related orbivirus are still associated with a significant impact on animal health and have important economic consequences worldwide. Our studies may contribute to the advance in orbivirus research and pave the way for the rapid development of new treatments, including vaccines.
Collapse
Affiliation(s)
- Sergio Utrilla-Trigo
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Luis Jiménez-Cabello
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Miguel Illescas-Amo
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Germán Andrés
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Gema Lorenzo
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Piet A. van Rijn
- Department of Virology, Wageningen Bioveterinary Research (WBVR), Lelystad, the Netherlands
- Department of Biochemistry, Centre for Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| | - Aitor Nogales
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Valdeolmos, Madrid, Spain
| |
Collapse
|
7
|
Londoño AF, Farner JM, Dillon M, Grab DJ, Kim Y, Scorpio DG, Dumler JS. Benidipine impairs innate immunity converting sublethal to lethal infections in a murine model of spotted fever rickettsiosis. PLoS Negl Trop Dis 2024; 18:e0011993. [PMID: 38408129 PMCID: PMC10919851 DOI: 10.1371/journal.pntd.0011993] [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: 09/07/2023] [Revised: 03/07/2024] [Accepted: 02/13/2024] [Indexed: 02/28/2024] Open
Abstract
Spotted fever group rickettsiae are tick-borne obligate intracellular bacteria that infect microvascular endothelial cells. Humans and mammalian infection results in endothelial cell barrier dysfunction and increased vascular permeability. We previously demonstrated that treatment of Rickettsia parkeri-infected cells with the calcium channel blocker benidipine significantly delayed vascular barrier permeability. Thus, we hypothesized that benidipine, known to be safe and effective for other clinical processes, could reduce rickettsia-induced vascular permeability in vivo in an animal model of spotted fever rickettsiosis. Based on liver, lung and brain vascular FITC-dextran extravasation studies, benidipine did not reliably impact vascular permeability. However, it precipitated a deleterious effect on responses to control sublethal R. parkeri infection. Animals treated with benidipine alone had no clinical signs or changes in histopathology and splenic immune cell distributions. Benidipine-treated infected animals had marked increases in tissue and blood bacterial loads, more extensive inflammatory histopathologic injury, and changes in splenic architecture and immune cell distributions potentially reflecting diminished Ca2+ signaling, reduced innate immune cell activation, and loss of rickettsial propagation control. Impaired T cell activation by R. parkeri antigen in the presence of benidipine was confirmed in vitro with the use of NKT cell hybridomas. The unexpected findings stand in stark contrast to recent discussions of the benefits of calcium channel blockers for viral infections and chronic infectious or inflammatory diseases. A role for calcium channel blockers in exacerbation of human rickettsiosis and acute inflammatory infections should be evaluated by a retrospective review of patient's outcomes and medications.
Collapse
Affiliation(s)
- Andrés F. Londoño
- Henry M. Jackson Foundation for Advancement of Military Medicine, Bethesda, Maryland, United States of America
- Department of Pathology, School of Medicine, Uniformed Services University, Bethesda, Maryland, United States of America
| | - Jennifer M. Farner
- Henry M. Jackson Foundation for Advancement of Military Medicine, Bethesda, Maryland, United States of America
- Emerging Infectious Disease Graduate Program, School of Medicine, Uniformed Services University, Bethesda, Maryland, United States of America
| | - Marlon Dillon
- Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dennis J. Grab
- Department of Pathology, School of Medicine, Uniformed Services University, Bethesda, Maryland, United States of America
| | - Yuri Kim
- Henry M. Jackson Foundation for Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Diana G. Scorpio
- Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - J. Stephen Dumler
- Department of Pathology, School of Medicine, Uniformed Services University, Bethesda, Maryland, United States of America
| |
Collapse
|
8
|
Islam MM, Alam MS, Liu Z, Khatun MS, Yusuf B, Hameed HMA, Tian X, Chhotaray C, Basnet R, Abraha H, Zhang X, Khan SA, Fang C, Li C, Hasan S, Tan S, Zhong N, Hu J, Zhang T. Molecular mechanisms of resistance and treatment efficacy of clofazimine and bedaquiline against Mycobacterium tuberculosis. Front Med (Lausanne) 2024; 10:1304857. [PMID: 38274444 PMCID: PMC10809401 DOI: 10.3389/fmed.2023.1304857] [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: 09/30/2023] [Accepted: 11/21/2023] [Indexed: 01/27/2024] Open
Abstract
Clofazimine (CFZ) and bedaquiline (BDQ) are currently used for the treatment of multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) strains. In recent years, adding CFZ and BDQ to tuberculosis (TB) drug regimens against MDR Mtb strains has significantly improved treatment results, but these improvements are threatened by the emergence of MDR and extensively drug-resistant (XDR) Mtb strains. Recently, CFZ and BDQ have attracted much attention for their strong clinical efficacy, although very little is known about the mechanisms of action, drug susceptibility test (DST), resistance mechanisms, cross-resistance, and pharmacokinetics of these two drugs. In this current review, we provide recent updates on the mechanisms of action, DST, associated mutations with individual resistance and cross-resistance, clinical efficacy, and pharmacokinetics of CFZ and BDQ against Mtb strains. Presently, known mechanisms of resistance for CFZ and/or BDQ include mutations within the Rv0678, pepQ, Rv1979c, and atpE genes. The cross-resistance between CFZ and BDQ may reduce available MDR-/XDR-TB treatment options. The use of CFZ and BDQ for treatment in the setting of limited DST could allow further spread of drug resistance. The DST and resistance knowledge are urgently needed where CFZ and BDQ resistance do emerge. Therefore, an in-depth understanding of clinical efficacy, DST, cross-resistance, and pharmacokinetics for CFZ and BDQ against Mtb can provide new ideas for improving treatment outcomes, reducing mortality, preventing drug resistance, and TB transmission. Along with this, it will also help to develop rapid molecular diagnostic tools as well as novel therapeutic drugs for TB.
Collapse
Affiliation(s)
- Md Mahmudul Islam
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Department of Microbiology, Shaheed Shamsuzzoha Institute of Biosciences, Affiliated with University of Rajshahi, Rajshahi, Bangladesh
| | - Md Shah Alam
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhiyong Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Mst Sumaia Khatun
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Buhari Yusuf
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - H. M. Adnan Hameed
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xirong Tian
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Chiranjibi Chhotaray
- Department of Medicine, Center for Emerging Pathogens, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Rajesh Basnet
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Haftay Abraha
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaofan Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shahzad Akbar Khan
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Laboratory of Pathology, Department of Pathobiology, University of Poonch Rawalakot, Azad Kashmir, Pakistan
| | - Cuiting Fang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Chunyu Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Sohel Hasan
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Shouyong Tan
- Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou, China
| | - Nanshan Zhong
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinxing Hu
- Guangzhou National Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
9
|
Xu H, Jian X, Wen Y, Xu M, Jin R, Wu X, Zhou F, Cao J, Xiao G, Peng K, Xie Y, Chen H, Zhang L. A nanoluciferase SFTSV for rapid screening antivirals and real-time visualization of virus infection in mice. EBioMedicine 2024; 99:104944. [PMID: 38176215 PMCID: PMC10806088 DOI: 10.1016/j.ebiom.2023.104944] [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: 04/08/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne pathogen that causes severe hemorrhagic fever in humans, but no FDA-approved specific antivirals or vaccines are available to treat or prevent SFTS. METHODS The plasmids construction and transfection were performed to generate the recombinant SFTSV harboring the nanoluciferase gene (SFTSV-Nluc). Immunostaining plaque assay was performed to measure viral titers, and DNA electrophoresis and Sanger sequencing were performed to evaluate the genetic stability. Luciferase assay and quantitative RT-PCR were performed to evaluate the efficacy of antivirals in vitro. Bioluminescence imaging, titration of virus from excised organs, hematology, and histopathology and immunohistochemistry were performed to evaluate the efficacy of antivirals in vivo. FINDINGS SFTSV-Nluc exhibited high genetic stability and replication kinetics similar to those of wild-type virus (SFTSVwt), then a rapid high-throughput screening system for identifying inhibitors to treat SFTS was developed, and a nucleoside analog, 4-FlU, was identified to effectively inhibit SFTSV in vitro. SFTSV-Nluc mimicked the replication characteristics and localization of SFTSVwt in counterpart model mice. Bioluminescence imaging of SFTSV-Nluc allowed real-time visualization and quantification of SFTSV replication in the mice. 4-FlU was demonstrated to inhibit the replication of SFTSV with more efficiency than T-705 and without obvious adverse effect in vivo. INTERPRETATION The high-throughput screening system based on SFTSV-Nluc for use in vitro and in vivo revealed that a safe and effective antiviral nucleoside analog, 4-FlU, may be a basis for the strategic treatment of SFTSV and other bunyavirus infections, paving the way for the discovery of antivirals. FUNDING This work was supported by grants from the National Key Research and Development Plan of China (2021YFC2300700 to L. Zhang, 2022YFC2303300 to L. Zhang), Strategic Priority Research Program of Chinese Academy of Sciences (XDB0490000 to L. Zhang), National Natural Science Foundation of China (31970165 to L. Zhang, U22A20379 to G. Xiao), the Science and Technology Commission of Shanghai Municipality (21S11903100 to Y. Xie), Hubei Natural Science Foundation for Distinguished Young Scholars (2022CFA099 to L. Zhang).
Collapse
Affiliation(s)
- Huan Xu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoqin Jian
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuxi Wen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mengwei Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fen Zhou
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Junyuan Cao
- Hubei Jiangxia Laboratory, Wuhan, 430200, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ke Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China; University of Chinese Academy of Sciences, Beijing, China.
| | | | - Hongbo Chen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China; University of Chinese Academy of Sciences, Beijing, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China.
| |
Collapse
|
10
|
Witwit H, Khafaji R, Salaniwal A, Kim AS, Cubitt B, Jackson N, Ye C, Weiss SR, Martinez-Sobrido L, de la Torre JC. Activation of Protein Kinase R (PKR) Plays a Pro-Viral Role in Mammarenavirus Infected Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570143. [PMID: 38106082 PMCID: PMC10723269 DOI: 10.1101/2023.12.05.570143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Many viruses, including mammarenaviruses, have evolved mechanisms to counteract different components of the host cell innate immunity, which is required to facilitate robust virus multiplication. The double strand (ds)RNA sensor protein kinase receptor (PKR) pathway plays a critical role in the cell antiviral response. Whether PKR can restrict the multiplication of the Old World mammarenavirus lymphocytic choriomeningitis virus (LCMV) and the mechanisms by which LCMV may counteract the antiviral functions of PKR have not yet been investigated. Here we present evidence that LCMV infection results in very limited levels of PKR activation, but LCMV multiplication is enhanced in the absence of PKR. In contrast, infection with a recombinant LCMV with a mutation affecting the 3'-5' exonuclease (ExoN) activity of the viral nucleoprotein (NP) resulted in robust PKR activation in the absence of detectable levels of dsRNA, which was associated with severely restricted virus multiplication that was alleviated in the absence of PKR. However, pharmacological inhibition of PKR activation resulted in reduced levels of LCMV multiplication. These findings uncovered a complex role of the PKR pathway in LCMV-infected cells involving both pro-and antiviral activities.
Collapse
Affiliation(s)
- Haydar Witwit
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Roaa Khafaji
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Arul Salaniwal
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Arthur S. Kim
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Beatrice Cubitt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Susan R Weiss
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | | | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
11
|
Guan J, Fan Y, Wang S, Zhou F. Functions of MAP3Ks in antiviral immunity. Immunol Res 2023; 71:814-832. [PMID: 37286768 PMCID: PMC10247270 DOI: 10.1007/s12026-023-09401-4] [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] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 06/01/2023] [Indexed: 06/09/2023]
Abstract
Immune signal transduction is crucial to the body's defense against viral infection. Recognition of pathogen-associated molecular patterns by pattern recognition receptors (PRRs) activates the transcription of interferon regulators and nuclear factor-κB (NF-κB); this promotes the release of interferons and inflammatory factors. Efficient regulation of type I interferon and NF-κB signaling by members of the mitogen-activated protein (MAP) kinase kinase kinase (MAP3K) family plays an important role in antiviral immunity. Elucidating the specific roles of MAP3K activation during viral infection is essential to develop effective antiviral therapies. In this review, we outline the specific regulatory mechanisms of MAP3Ks in antiviral immunity and discuss the feasibility of targeting MAP3Ks for the treatment of virus-induced diseases.
Collapse
Affiliation(s)
- Jizhong Guan
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China
| | - Yao Fan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Shuai Wang
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
12
|
Xu M, Xu H, Wan W, Jian X, Jin R, Wang L, Wang J, Xiao G, Zhang L, Chen H, Wen Y. PDIA4 Is a Host Factor Important for Lymphocytic Choriomeningitis Virus Infection. Viruses 2023; 15:2343. [PMID: 38140584 PMCID: PMC10747894 DOI: 10.3390/v15122343] [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: 10/18/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Mammalian arenaviruses are rodent-borne zoonotic viruses, some of which can cause fatal hemorrhagic diseases in humans. The first discovered arenavirus, lymphocytic choriomeningitis virus (LCMV), has a worldwide distribution and can be fatal for transplant recipients. However, no FDA-approved drugs or vaccines are currently available. In this study, using a quantitative proteomic analysis, we identified a variety of host factors that could be needed for LCMV infection, among which we found that protein disulfide isomerase A4 (PDIA4), a downstream factor of endoplasmic reticulum stress (ERS), is important for LCMV infection. Biochemical analysis revealed that LCMV glycoprotein was the main viral component accounting for PDIA4 upregulation. The inhibition of ATF6-mediated ERS could prevent the upregulation of PDIA4 that was stimulated by LCMV infection. We further found that PDIA4 can affect the LCMV viral RNA synthesis processes and release. In summary, we conclude that PDIA4 could be a new target for antiviral drugs against LCMV.
Collapse
Affiliation(s)
- Mengwei Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; (M.X.); (W.W.); (X.J.); (G.X.); (L.Z.)
| | - Huan Xu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (H.X.); (R.J.); (L.W.)
| | - Weiwei Wan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; (M.X.); (W.W.); (X.J.); (G.X.); (L.Z.)
| | - Xiaoqin Jian
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; (M.X.); (W.W.); (X.J.); (G.X.); (L.Z.)
- University of Chinese Academy of Sciences, Beijing 100000, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (H.X.); (R.J.); (L.W.)
| | - Lin Wang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (H.X.); (R.J.); (L.W.)
| | - Jingshi Wang
- Department of Hematology, Beijing Friendship Hospital, Capital Medical University, Beijing 100000, China;
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; (M.X.); (W.W.); (X.J.); (G.X.); (L.Z.)
- University of Chinese Academy of Sciences, Beijing 100000, China
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; (M.X.); (W.W.); (X.J.); (G.X.); (L.Z.)
- University of Chinese Academy of Sciences, Beijing 100000, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| | - Hongbo Chen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (H.X.); (R.J.); (L.W.)
| | - Yuxi Wen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (H.X.); (R.J.); (L.W.)
| |
Collapse
|
13
|
LaLone V, Smith D, Diaz-Espinosa J, Rosania GR. Quantitative Raman chemical imaging of intracellular drug-membrane aggregates and small molecule drug precipitates in cytoplasmic organelles. Adv Drug Deliv Rev 2023; 202:115107. [PMID: 37769851 PMCID: PMC10841539 DOI: 10.1016/j.addr.2023.115107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Raman confocal microscopes have been used to visualize the distribution of small molecule drugs within different subcellular compartments. This visualization allows the discovery, characterization, and detailed analysis of the molecular transport phenomena underpinning the Volume of Distribution - a key parameter governing the systemic pharmacokinetics of small molecule drugs. In the specific case of lipophilic small molecules with large Volumes of Distribution, chemical imaging studies using Raman confocal microscopes have revealed how weakly basic, poorly soluble drug molecules can accumulate inside cells by forming stable, supramolecular complexes in association with cytoplasmic membranes or by precipitating out within organelles. To study the self-assembly and function of the resulting intracellular drug inclusions, Raman chemical imaging methods have been developed to measure and map the mass, concentration, and ionization state of drug molecules at a microscopic, subcellular level. Beyond the field of drug delivery, Raman chemical imaging techniques relevant to the study of microscopic drug precipitates and drug-lipid complexes which form inside cells are also being developed by researchers with seemingly unrelated scientific interests. Highlighting advances in data acquisition, calibration methods, and computational data management and analysis tools, this review will cover a decade of technological developments that enable the conversion of spectral signals obtained from Raman confocal microscopes into new discoveries and information about previously unknown, concentrative drug transport pathways driven by soluble-to-insoluble phase transitions occurring within the cytoplasmic organelles of eukaryotic cells.
Collapse
Affiliation(s)
- Vernon LaLone
- Cambium Analytica Research Laboratories, Traverse City, MI, United States
| | - Doug Smith
- Cambium Analytica Research Laboratories, Traverse City, MI, United States
| | - Jennifer Diaz-Espinosa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Gus R Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
14
|
Xu J, Koval A, Katanaev VL. Clofazimine: A journey of a drug. Biomed Pharmacother 2023; 167:115539. [PMID: 37742606 DOI: 10.1016/j.biopha.2023.115539] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/12/2023] [Accepted: 09/17/2023] [Indexed: 09/26/2023] Open
Abstract
Among different strategies to develop novel therapies, drug repositioning (aka repurposing) aims at identifying new uses of an already approved or investigational drug. This approach has the advantages of availability of the extensive pre-existing knowledge of the drug's safety, pharmacology and toxicology, manufacturing and formulation. It provides advantages to the risk-versus-rewards trade-off as compared to the costly and time-consuming de novo drug discovery process. Clofazimine, a red-colored synthetic derivative of riminophenazines initially isolated from lichens, was first synthesized in the 1950 s, and passed through several phases of repositioning in its history as a drug. Being initially developed as an anti-tuberculosis treatment, it was repurposed for the treatment of leprosy, prior to re-repositioning for the treatment of multidrug-resistant tuberculosis and other infections. Since 1990 s, reports on the anticancer properties of clofazimine, both in vitro and in vivo, started to appear. Among the diverse mechanisms of action proposed, the activity of clofazimine as a specific inhibitor of the oncogenic Wnt signaling pathway has recently emerged as the promising targeting mechanism of the drug against breast, colon, liver, and other forms of cancer. Seventy years after the initial discovery, clofazimine's journey as a drug finding new applications continues, serving as a colorful illustration of drug repurposing in modern pharmacology.
Collapse
Affiliation(s)
- Jiabin Xu
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Vladimir L Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok, Russia.
| |
Collapse
|
15
|
Hamele CE, Spurrier MA, Leonard RA, Heaton NS. Segmented, Negative-Sense RNA Viruses of Humans: Genetic Systems and Experimental Uses of Reporter Strains. Annu Rev Virol 2023; 10:261-282. [PMID: 37774125 PMCID: PMC10795101 DOI: 10.1146/annurev-virology-111821-120445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
Negative-stranded RNA viruses are a large group of viruses that encode their genomes in RNA across multiple segments in an orientation antisense to messenger RNA. Their members infect broad ranges of hosts, and there are a number of notable human pathogens. Here, we examine the development of reverse genetic systems as applied to these virus families, emphasizing conserved approaches illustrated by some of the prominent members that cause significant human disease. We also describe the utility of their genetic systems in the development of reporter strains of the viruses and some biological insights made possible by their use. To conclude the review, we highlight some possible future uses of reporter viruses that not only will increase our basic understanding of how these viruses replicate and cause disease but also could inform the development of new approaches to therapeutically intervene.
Collapse
Affiliation(s)
- Cait E Hamele
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA;
| | - M Ariel Spurrier
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA;
| | - Rebecca A Leonard
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA;
| | - Nicholas S Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA;
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
16
|
Shen S, Zhang Y, Yin Z, Zhu Q, Zhang J, Wang T, Fang Y, Wu X, Bai Y, Dai S, Liu X, Jin J, Tang S, Liu J, Wang M, Guo Y, Deng F. Antiviral activity and mechanism of the antifungal drug, anidulafungin, suggesting its potential to promote treatment of viral diseases. BMC Med 2022; 20:359. [PMID: 36266654 PMCID: PMC9585728 DOI: 10.1186/s12916-022-02558-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The severe fever with thrombocytopenia syndrome disease (SFTS), caused by the novel tick-borne SFTS virus (SFTSV), was listed among the top 10 priority infectious disease by World Health Organization due to the high fatality rate of 5-30% and the lack of effective antiviral drugs and vaccines and therefore raised the urgent need to develop effective anti-SFTSV drugs to improve disease treatment. METHODS The antiviral drugs to inhibit SFTSV infection were identified by screening the library containing 1340 FDA-approved drugs using the SFTSV infection assays in vitro. The inhibitory effect on virus entry and the process of clathrin-mediated endocytosis under different drug doses was evaluated based on infection assays by qRT-PCR to determine intracellular viral copies, by Western blot to characterize viral protein expression in cells, and by immunofluorescence assays (IFAs) to determine virus infection efficiencies. The therapeutic effect was investigated in type I interferon receptor defective A129 mice in vivo with SFTSV infection, from which lesions and infection in tissues caused by SFTSV infection were assessed by H&E staining and immunohistochemical analysis. RESULTS Six drugs were identified as exerting inhibitory effects against SFTSV infection, of which anidulafungin, an antifungal drug of the echinocandin family, has a strong inhibitory effect on SFTSV entry. It suppresses SFTSV internalization by impairing the late endosome maturation and decreasing virus fusion with the membrane. SFTSV-infected A129 mice had relieving symptoms, reduced tissue lesions, and improved disease outcomes following anidulafungin treatment. Moreover, anidulafungin exerts an antiviral effect in inhibiting the entry of other viruses including SARS-CoV-2, SFTSV-related Guertu virus and Heartland virus, Crimean-Congo hemorrhagic fever virus, Zika virus, and Herpes simplex virus 1. CONCLUSIONS The results demonstrated that the antifungal drug, anidulafungin, could effectively inhibit virus infection by interfering with virus entry, suggesting it may be utilized for the clinical treatment of infectious viral diseases, in addition to its FDA-approved use as an antifungal. The findings also suggested to further evaluate the anti-viral effects of echinocandins and their clinical importance for patients with infection of viruses, which may promote therapeutic strategies as well as treatments and improve outcomes pertaining to various viral and fungal diseases.
Collapse
Affiliation(s)
- Shu Shen
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Yaxian Zhang
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China.,State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Zhiyun Yin
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China.,State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Qiong Zhu
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Jingyuan Zhang
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Tiantian Wang
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Yaohui Fang
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Xiaoli Wu
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Yuan Bai
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Shiyu Dai
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Xijia Liu
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Jiayin Jin
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Shuang Tang
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Jia Liu
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Manli Wang
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China
| | - Yu Guo
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, China.,College of Life Science, Nankai University, Tianjin, 300350, China
| | - Fei Deng
- State Key Laboratory of virology and National Virus Resource Center, Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan 44#, Wuchang District, Wuhan, 430071, Hubei, China.
| |
Collapse
|
17
|
Wen Y, Xu H, Wan W, Shang W, Jin R, Zhou F, Mei H, Wang J, Xiao G, Chen H, Wu X, Zhang L. Visualizing lymphocytic choriomeningitis virus infection in cells and living mice. iScience 2022; 25:105090. [PMID: 36185356 PMCID: PMC9519613 DOI: 10.1016/j.isci.2022.105090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Mammarenavirus are a large family of enveloped negative-strand RNA viruses that include several agents responsible for severe hemorrhagic fevers. Until now, no FDA-licensed drug has been admitted for treating an arenavirus infection, and only few effective anti-arenavirus drugs have been tested in vivo. In this work, we designed a recombinant reporter arenavirus lymphocytic choriomeningitis virus that stably expressed nanoluciferase (LCMV-Nluc). The LCMV-Nluc was proved to share similar biological properties with wild-type LCMV and the Nluc intensity reliably reflected viral replication both in vitro and in vivo. Replication of the Nluc-encoding virus in living mice can be visualized by real-time bioluminescent imaging, and bioluminescence can be detected in a variety of organs of infected mice. This work provides a novel approach that enables real-time study of the arenavirus infection and is a convenient and valuable tool for screening of compounds that are active against arenaviruses in vitro and in living mice. LCMV-Nluc was constructed and shared similar biological properties with LCMV-WT Replication of the LCMV-Nluc can be visualized by real-time bioluminescent imaging LCMV-Nluc is a valuable tool for screening antiviral compounds in vitro LCMV-Nluc is successfully applied for screening antiviral compounds in vivo
Collapse
Affiliation(s)
- Yuxi Wen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huan Xu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiwei Wan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Weijuan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fen Zhou
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heng Mei
- Institute of Haematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Clinical and Research Centre of Thrombosis and Haemostasis, Wuhan, China
| | - Jingshi Wang
- Department of Hematology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongbo Chen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Hubei Jiangxia Laboratory,Wuhan 430000, China
| |
Collapse
|
18
|
Factors Associated With COVID-19 Vaccine Response in Transplant Recipients: A Systematic Review and Meta-analysis. Transplantation 2022; 106:2068-2075. [PMID: 35761439 PMCID: PMC9521391 DOI: 10.1097/tp.0000000000004256] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The rapid development and universal access to vaccines represent a milestone in combating the coronavirus disease 2019 (COVID-19) pandemic. However, there are major concerns about vaccine response in immunocompromised populations in particular transplant recipients. In the present study, we aim to comprehensively assess the humoral response to COVID-19 vaccination in both orthotopic organ transplant and allogeneic hematopoietic stem cell transplant recipients. METHODS We performed a systematic review and meta-analysis of 96 studies that met inclusion criteria. RESULTS The pooled rates of seroconversion were 49% (95% confidence interval [CI], 43%-55%) in transplant recipients and 99% (95% CI, 99%-99%) in healthy controls after the second dose of vaccine. The pooled rate was 56% (95% CI, 49%-63%) in transplant recipients after the third dose. Immunosuppressive medication is the most prominent risk factor associated with seroconversion failure, but different immunosuppressive regimens are associated with differential outcomes in this respect. Calcineurin inhibitors, steroids, or mycophenolate mofetil/mycophenolic acid are associated with an increased risk of seroconversion failure, whereas azathioprine or mammalian target of rapamycin inhibitors do not. Advanced age, short interval from receiving the vaccine to the time of transplantation, or comorbidities confers a higher risk for seroconversion failure. CONCLUSIONS Transplant recipients compared with the general population have much lower rates of seroconversion upon receiving COVID-19 vaccines. Immunosuppressants are the most prominent factors associated with seroconversion, although different types may have differential effects.
Collapse
|
19
|
Fénéant L, Leske A, Günther K, Groseth A. Generation of Reporter-Expressing New World Arenaviruses: A Systematic Comparison. Viruses 2022; 14:v14071563. [PMID: 35891543 PMCID: PMC9317149 DOI: 10.3390/v14071563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 02/01/2023] Open
Abstract
Replication-competent reporter-expressing viruses are crucial tools in molecular virology with applications that range from antiviral screening to live-cell imaging of protein spatiotemporal dynamics. However, there is currently little information available regarding viable strategies to develop reporter-expressing arenaviruses. To address this, we used Tacaribe virus (TCRV), an apathogenic BSL2 arenavirus, to assess the feasibility of different reporter expression approaches. We first generated trisegmented TCRV viruses with either the glycoprotein (GP) or nucleoprotein (NP) replaced by a reporter (GFP, mCherry, or nanoluciferase). These viruses were all viable, but showed marked differences in brightness and attenuation. Next, we generated terminal fusions with each of the TCRV proteins (i.e., NP, GP, polymerase (L), matrix protein (Z)) either with or without a T2A self-cleavage site. We tested both the function of the reporter-fused proteins alone, and the viability of corresponding recombinant TCRVs. We successfully rescued viruses with both direct and cleavable reporter fusions at the C-terminus of Z, as well as cleavable N-terminal fusions with NP. These viruses all displayed detectable reporter activity, but were also moderately attenuated. Finally, reporter proteins were inserted into a flexible hinge region within L. These viruses were also viable and showed moderate attenuation; however, reporter expression was only detectable for the luminescent virus. These strategies provide an exciting range of new tools for research into the molecular biology of TCRV that can likely also be adapted to other arenaviruses.
Collapse
|
20
|
Qu N, Hui Z, Shen Z, Kan C, Hou N, Sun X, Han F. Thyroid Cancer and COVID-19: Prospects for Therapeutic Approaches and Drug Development. Front Endocrinol (Lausanne) 2022; 13:873027. [PMID: 35600591 PMCID: PMC9114699 DOI: 10.3389/fendo.2022.873027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/04/2022] [Indexed: 02/05/2023] Open
Abstract
Thyroid cancer is the most prevalent endocrine malignancy and the reported incidence of thyroid cancer has continued to increase in recent years. Since 2019, coronavirus disease 2019 (COVID-19) has been spreading worldwide in a global pandemic. COVID-19 aggravates primary illnesses and affects disease management; relevant changes include delayed diagnosis and treatment. The thyroid is an endocrine organ that is susceptible to autoimmune attack; thus, thyroid cancer after COVID-19 has gradually attracted attention. Whether COVID-19 affects the diagnosis and treatment of thyroid cancer has also attracted the attention of many researchers. This review examines the literature regarding the influence of COVID-19 on the pathogenesis, diagnosis, and treatment of thyroid cancer; it also focuses on drug therapies to promote research into strategies for improving therapy and management in thyroid cancer patients with COVID-19.
Collapse
Affiliation(s)
- Na Qu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zongguang Hui
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zhixin Shen
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
21
|
Li TW, Kenney AD, Park JG, Fiches GN, Liu H, Zhou D, Biswas A, Zhao W, Que J, Santoso N, Martinez-Sobrido L, Yount JS, Zhu J. SARS-CoV-2 Nsp14 protein associates with IMPDH2 and activates NF-κB signaling. Front Immunol 2022; 13:1007089. [PMID: 36177032 PMCID: PMC9513374 DOI: 10.3389/fimmu.2022.1007089] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to NF-κB activation and induction of pro-inflammatory cytokines, though the underlying mechanism for this activation is not fully understood. Our results reveal that the SARS-CoV-2 Nsp14 protein contributes to the viral activation of NF-κB signaling. Nsp14 caused the nuclear translocation of NF-κB p65. Nsp14 induced the upregulation of IL-6 and IL-8, which also occurred in SARS-CoV-2 infected cells. IL-8 upregulation was further confirmed in lung tissue samples from COVID-19 patients. A previous proteomic screen identified the putative interaction of Nsp14 with host Inosine-5'-monophosphate dehydrogenase 2 (IMPDH2), which is known to regulate NF-κB signaling. We confirmed the Nsp14-IMPDH2 protein interaction and identified that IMPDH2 knockdown or chemical inhibition using ribavirin (RIB) and mycophenolic acid (MPA) abolishes Nsp14- mediated NF-κB activation and cytokine induction. Furthermore, IMPDH2 inhibitors (RIB, MPA) or NF-κB inhibitors (bortezomib, BAY 11-7082) restricted SARS-CoV-2 infection, indicating that IMPDH2-mediated activation of NF-κB signaling is beneficial to viral replication. Overall, our results identify a novel role of SARS-CoV-2 Nsp14 in inducing NF-κB activation through IMPDH2 to promote viral infection.
Collapse
Affiliation(s)
- Tai-Wei Li
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Adam D. Kenney
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jun-Gyu Park
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Guillaume N. Fiches
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Helu Liu
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Dawei Zhou
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Ayan Biswas
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Weiqiang Zhao
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Netty Santoso
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | | | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jian Zhu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- *Correspondence: Jian Zhu,
| |
Collapse
|
22
|
Dunne S, Willmer AR, Swanson R, Almeida D, Ammerman NC, Stringer KA, Capparelli EV, Rosania GR. Quantitative Analysis of the Phase Transition Mechanism Underpinning the Systemic Self-Assembly of a Mechanopharmaceutical Device. Pharmaceutics 2021; 14:15. [PMID: 35056910 PMCID: PMC8780429 DOI: 10.3390/pharmaceutics14010015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 01/01/2023] Open
Abstract
Clofazimine (CFZ) is a poorly soluble, weakly basic, small molecule antibiotic clinically used to treat leprosy and is now in clinical trials as a treatment for multidrug resistant tuberculosis and COVID-19. CFZ exhibits complex, context-dependent pharmacokinetics that are characterized by an increasing half-life in long term treatment regimens. The systemic pharmacokinetics of CFZ have been previously represented by a nonlinear, 2-compartment model incorporating an expanding volume of distribution. This expansion reflects the soluble-to-insoluble phase transition that the drug undergoes as it precipitates out and accumulates within macrophages disseminated throughout the organism. Using mice as a model organism, we studied the mechanistic underpinnings of this increasing half-life and how the systemic pharmacokinetics of CFZ are altered with continued dosing. To this end, M. tuberculosis infection status and multiple dosing schemes were studied alongside a parameter sensitivity analysis (PSA) to further understanding of systemic drug distribution. Parameter values governing the sigmoidal expansion function that captures the phase transition were methodically varied, and in turn, the systemic concentrations of the drug were calculated and compared to the experimentally measured concentrations of drug in serum and spleen. The resulting amounts of drug sequestered were dependent on the total mass of CFZ administered and the duration of drug loading. This phenomenon can be captured by altering three different parameters of an expansion function corresponding to key biological determinants responsible for the precipitation and the accumulation of the insoluble drug mass in macrophages. Through this analysis of the context dependent pharmacokinetics of CFZ, a predictive framework for projecting the systemic distribution and self-assembly of precipitated drug complexes as intracellular mechanopharmaceutical devices of this and other drugs exhibiting similarly complex pharmacokinetics can be constructed.
Collapse
Affiliation(s)
- Steven Dunne
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Andrew R. Willmer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Rosemary Swanson
- Johns Hopkins Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.S.); (D.A.); (N.C.A.)
| | - Deepak Almeida
- Johns Hopkins Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.S.); (D.A.); (N.C.A.)
| | - Nicole C. Ammerman
- Johns Hopkins Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.S.); (D.A.); (N.C.A.)
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Kathleen A. Stringer
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Edmund V. Capparelli
- Department of Pediatrics, Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, CA 92093, USA;
| | - Gus R. Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
23
|
Yin Z, Wong STC. Artificial intelligence unifies knowledge and actions in drug repositioning. Emerg Top Life Sci 2021; 5:803-813. [PMID: 34881780 PMCID: PMC8923082 DOI: 10.1042/etls20210223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022]
Abstract
Drug repositioning aims to reuse existing drugs, shelved drugs, or drug candidates that failed clinical trials for other medical indications. Its attraction is sprung from the reduction in risk associated with safety testing of new medications and the time to get a known drug into the clinics. Artificial Intelligence (AI) has been recently pursued to speed up drug repositioning and discovery. The essence of AI in drug repositioning is to unify the knowledge and actions, i.e. incorporating real-world and experimental data to map out the best way forward to identify effective therapeutics against a disease. In this review, we share positive expectations for the evolution of AI and drug repositioning and summarize the role of AI in several methods of drug repositioning.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center and Ting Tsung & Wei Fong Chao Center for BRAIN, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, U.S.A
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center and Ting Tsung & Wei Fong Chao Center for BRAIN, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, U.S.A
| |
Collapse
|
24
|
White JM, Schiffer JT, Bender Ignacio RA, Xu S, Kainov D, Ianevski A, Aittokallio T, Frieman M, Olinger GG, Polyak SJ. Drug Combinations as a First Line of Defense against Coronaviruses and Other Emerging Viruses. mBio 2021; 12:e0334721. [PMID: 34933447 PMCID: PMC8689562 DOI: 10.1128/mbio.03347-21] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The world was unprepared for coronavirus disease 2019 (COVID-19) and remains ill-equipped for future pandemics. While unprecedented strides have been made developing vaccines and treatments for COVID-19, there remains a need for highly effective and widely available regimens for ambulatory use for novel coronaviruses and other viral pathogens. We posit that a priority is to develop pan-family drug cocktails to enhance potency, limit toxicity, and avoid drug resistance. We urge cocktail development for all viruses with pandemic potential both in the short term (<1 to 2 years) and longer term with pairs of drugs in advanced clinical testing or repurposed agents approved for other indications. While significant efforts were launched against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in vitro and in the clinic, many studies employed solo drugs and had disappointing results. Here, we review drug combination studies against SARS-CoV-2 and other viruses and introduce a model-driven approach to assess drug pairs with the highest likelihood of clinical efficacy. Where component agents lack sufficient potency, we advocate for synergistic combinations to achieve therapeutic levels. We also discuss issues that stymied therapeutic progress against COVID-19, including testing of agents with low likelihood of efficacy late in clinical disease and lack of focus on developing virologic surrogate endpoints. There is a need to expedite efficient clinical trials testing drug combinations that could be taken at home by recently infected individuals and exposed contacts as early as possible during the next pandemic, whether caused by a coronavirus or another viral pathogen. The approach herein represents a proactive plan for global viral pandemic preparedness.
Collapse
Affiliation(s)
- Judith M. White
- University of Virginia, Department of Cell Biology, Charlottesville, Virginia, USA
- University of Virginia, Department of Microbiology, Charlottesville, Virginia, USA
| | - Joshua T. Schiffer
- University of Washington, Division of Allergy and Infectious Diseases, Seattle, Washington, USA
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Rachel A. Bender Ignacio
- University of Washington, Division of Allergy and Infectious Diseases, Seattle, Washington, USA
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Shuang Xu
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Institute of Technology, University of Tartu, Tartu, Estonia
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
- Oslo Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, Oslo, Norway
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Matthew Frieman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Stephen J. Polyak
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
25
|
Siminea N, Popescu V, Sanchez Martin JA, Florea D, Gavril G, Gheorghe AM, Iţcuş C, Kanhaiya K, Pacioglu O, Popa IL, Trandafir R, Tusa MI, Sidoroff M, Păun M, Czeizler E, Păun A, Petre I. Network analytics for drug repurposing in COVID-19. Brief Bioinform 2021; 23:6447433. [PMID: 34864885 PMCID: PMC8690228 DOI: 10.1093/bib/bbab490] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/06/2021] [Accepted: 10/25/2021] [Indexed: 01/08/2023] Open
Abstract
To better understand the potential of drug repurposing in COVID-19, we analyzed control strategies over essential host factors for SARS-CoV-2 infection. We constructed comprehensive directed protein–protein interaction (PPI) networks integrating the top-ranked host factors, the drug target proteins and directed PPI data. We analyzed the networks to identify drug targets and combinations thereof that offer efficient control over the host factors. We validated our findings against clinical studies data and bioinformatics studies. Our method offers a new insight into the molecular details of the disease and into potentially new therapy targets for it. Our approach for drug repurposing is significant beyond COVID-19 and may be applied also to other diseases.
Collapse
Affiliation(s)
- Nicoleta Siminea
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania.,Faculty of Mathematics and Computer Science, University of Bucharest, 14 Academiei, 010014, Romania
| | - Victor Popescu
- Department of Information Technologies, Åbo Akademi University, 3 Tuomiokirkontori, 20500, Finland
| | - Jose Angel Sanchez Martin
- Department of Computer Science, Technical University of Madrid, 7 Calle Ramiro de Maeztu, 28040, Spain
| | - Daniela Florea
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Georgiana Gavril
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Ana-Maria Gheorghe
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Corina Iţcuş
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Krishna Kanhaiya
- Department of Information Technologies, Åbo Akademi University, 3 Tuomiokirkontori, 20500, Finland
| | - Octavian Pacioglu
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Ioana Laura Popa
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Romica Trandafir
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Maria Iris Tusa
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Manuela Sidoroff
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Mihaela Păun
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania.,Faculty of Administration and Business, University of Bucharest, 4-12 Regina Elisabeta Boulevard, 030018, Romania
| | - Eugen Czeizler
- Department of Information Technologies, Åbo Akademi University, 3 Tuomiokirkontori, 20500, Finland.,Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| | - Andrei Păun
- Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania.,Faculty of Mathematics and Computer Science, University of Bucharest, 14 Academiei, 010014, Romania
| | - Ion Petre
- Department of Mathematics and Statistics, University of Turku, 5 Vesilinnantie, 20014, Finland.,Bioinformatics, National Institute of Research and Development for Biological Sciences, 296 Splaiul Independenţei, 060031, Romania
| |
Collapse
|
26
|
Loas G, Le Corre P. Update on Functional Inhibitors of Acid Sphingomyelinase (FIASMAs) in SARS-CoV-2 Infection. Pharmaceuticals (Basel) 2021; 14:691. [PMID: 34358117 PMCID: PMC8308787 DOI: 10.3390/ph14070691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
The SARS-CoV-2 outbreak is characterized by the need of the search for curative drugs for treatment. In this paper, we present an update of knowledge about the interest of the functional inhibitors of acid sphingomyelinase (FIASMAs) in SARS-CoV-2 infection. Forty-nine FIASMAs have been suggested in the treatment of SARS-CoV-2 infection using in silico, in vitro or in vivo studies. Further studies using large-sized, randomized and double-blinded controlled clinical trials are needed to evaluate FIASMAs in SARS-CoV-2 infection as off-label therapy.
Collapse
Affiliation(s)
- Gwenolé Loas
- Department of Psychiatry, Hôpital Erasme, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
- Research Unit (ULB 266), Hôpital Erasme, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - Pascal Le Corre
- Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU de Rennes, 35033 Rennes, France;
- Irset (Institut de Recherche en Santé, Environnement et Travail)-Inserm UMR 1085, University of Rennes, CHU Rennes, INSERM, EHESP, 35000 Rennes, France
- Laboratoire de Biopharmacie et Pharmacie Clinique, Faculté de Pharmacie, Université de Rennes 1, 35043 Rennes, France
| |
Collapse
|
27
|
Cicka D, Sukhatme VP. Available drugs and supplements for rapid deployment for treatment of COVID-19. J Mol Cell Biol 2021; 13:232-236. [PMID: 33493301 PMCID: PMC7928750 DOI: 10.1093/jmcb/mjab002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/09/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022] Open
Abstract
Abstract
Effective treatment for COVID-19 remains elusive, though urgently needed in the current pandemic. Repurposing marketed therapies may be an effective strategy for finding treatments quickly and, recently, in vitro and clinical testing of such therapies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has skyrocketed. However, not all marketed drugs showing in vitro efficacy could achieve therapeutic concentrations in humans and discernment of drugs that have favorable pharmacokinetic properties can save time and resources for future studies. Here, we compile marketed therapies, including supplements, having anti-viral activity with in vitro, in vivo, and/or clinical data against α and β coronaviruses into tables, alongside their pharmacokinetic properties. We point to several drugs or supplements available for immediate repurposing because they have achievable blood concentrations in humans well above their inhibitory concentrations against coronaviruses. This compilation may contribute to the implementation of rapid future studies by narrowing the vast number of marketed drugs reported for potential efficacy against SARS-CoV-2 on the basis of their pharmacokinetic properties and published coronavirus data.
Collapse
Affiliation(s)
- Danielle Cicka
- Department of Pharmacology and Chemical Biology,
Emory University School of Medicine, Atlanta, GA, USA
| | - Vikas P Sukhatme
- Department of Medicine, Department of Hematology and
Medical Oncology, Morningside Center for Innovative and Affordable Medicine,
Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
28
|
Goldman JD, Robinson PC, Uldrick TS, Ljungman P. COVID-19 in immunocompromised populations: implications for prognosis and repurposing of immunotherapies. J Immunother Cancer 2021; 9:e002630. [PMID: 34117116 PMCID: PMC8206176 DOI: 10.1136/jitc-2021-002630] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 is the virus responsible for the COVID-19 pandemic. COVID-19 has highly variable disease severity and a bimodal course characterized by acute respiratory viral infection followed by hyperinflammation in a subset of patients with severe disease. This immune dysregulation is characterized by lymphocytopenia, elevated levels of plasma cytokines and proliferative and exhausted T cells, among other dysfunctional cell types. Immunocompromised persons often fare worse in the context of acute respiratory infections, but preliminary data suggest this may not hold true for COVID-19. In this review, we explore the effect of SARS-CoV-2 infection on mortality in four populations with distinct forms of immunocompromise: (1) persons with hematological malignancies (HM) and hematopoietic stem cell transplant (HCT) recipients; (2) solid organ transplant recipients (SOTRs); (3) persons with rheumatological diseases; and (4) persons living with HIV (PLWH). For each population, key immunological defects are described and how these relate to the immune dysregulation in COVID-19. Next, outcomes including mortality after SARS-CoV-2 infection are described for each population, giving comparisons to the general population of age-matched and comorbidity-matched controls. In these four populations, iatrogenic or disease-related immunosuppression is not clearly associated with poor prognosis in HM, HCT, SOTR, rheumatological diseases, or HIV. However, certain individual immunosuppressants or disease states may be associated with harmful or beneficial effects, including harm from severe CD4 lymphocytopenia in PLWH and possible benefit to the calcineurin inhibitor ciclosporin in SOTRs, or tumor necrosis factor-α inhibitors in persons with rheumatic diseases. Lastly, insights gained from clinical and translational studies are explored as to the relevance for repurposing of immunosuppressive host-directed therapies for the treatment of hyperinflammation in COVID-19 in the general population.
Collapse
Affiliation(s)
- Jason D Goldman
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, Washington, USA
- Providence St. Joseph Health, Renton, Washington, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Philip C Robinson
- The University of Queensland Faculty of Medicine, Herston, Queensland, Australia
- Metro North Hospital and Health Service, Royal Brisbane and Woman's Hospital Health Service District, Herston, Queensland, Australia
| | - Thomas S Uldrick
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Per Ljungman
- Department. of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, Karolinska Comprehensive Cancer Center, Stockholm, Sweden
- Division of Hematology, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Li T, Kenney AD, Liu H, Fiches GN, Zhou D, Biswas A, Que J, Santoso N, Yount JS, Zhu J. SARS-CoV-2 Nsp14 activates NF-κB signaling and induces IL-8 upregulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.26.445787. [PMID: 34075374 PMCID: PMC8168382 DOI: 10.1101/2021.05.26.445787] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to NF-κB activation and induction of pro-inflammatory cytokines, though the underlying mechanism for this activation is not fully understood. Our results reveal that the SARS-CoV-2 Nsp14 protein contributes to the viral activation of NF-κB signaling. Nsp14 caused the nuclear translocation of NF-κB p65. Nsp14 induced the upregulation of IL-6 and IL-8, which also occurred in SARS-CoV-2 infected cells. IL-8 upregulation was further confirmed in lung tissue samples from COVID-19 patients. A previous proteomic screen identified the putative interaction of Nsp14 with host Inosine-5'-monophosphate dehydrogenase 2 (IMPDH2) protein, which is known to regulate NF-κB signaling. We confirmed the Nsp14-IMPDH2 protein interaction and found that IMPDH2 knockdown or chemical inhibition using ribavirin (RIB) and mycophenolic acid (MPA) abolishes Nsp14-mediated NF-κB activation and cytokine induction. Furthermore, IMDPH2 inhibitors (RIB, MPA) efficiently blocked SARS-CoV-2 infection, indicating that IMDPH2, and possibly NF-κB signaling, is beneficial to viral replication. Overall, our results identify a novel role of SARS-CoV-2 Nsp14 in causing the activation of NF-κB.
Collapse
Affiliation(s)
- Taiwei Li
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Adam D. Kenney
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Helu Liu
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Guillaume N. Fiches
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Dawei Zhou
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ayan Biswas
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Netty Santoso
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jian Zhu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
30
|
Vilibic-Cavlek T, Savic V, Ferenc T, Mrzljak A, Barbic L, Bogdanic M, Stevanovic V, Tabain I, Ferencak I, Zidovec-Lepej S. Lymphocytic Choriomeningitis-Emerging Trends of a Neglected Virus: A Narrative Review. Trop Med Infect Dis 2021; 6:88. [PMID: 34070581 PMCID: PMC8163193 DOI: 10.3390/tropicalmed6020088] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Lymphocytic choriomeningitis virus (LCMV) is a neglected rodent-borne zoonotic virus distributed worldwide. Since serologic assays are limited to several laboratories, the disease has been underreported, often making it difficult to determine incidence and seroprevalence rates. Although human clinical cases are rarely recorded, LCMV remains an important cause of meningitis in humans. In addition, a fatal donor-derived LCMV infection in several clusters of solid organ transplant recipients further highlighted a pathogenic potential and clinical significance of this virus. In the transplant populations, abnormalities of the central nervous system were also found, but were overshadowed by the systemic illness resembling the Lassa hemorrhagic fever. LCMV is also an emerging fetal teratogen. Hydrocephalus, periventricular calcifications and chorioretinitis are the predominant characteristics of congenital LCMV infection, occurring in 87.5% of cases. Mortality in congenitally infected children is about 35%, while 70% of them show long-term neurologic sequelae. Clinicians should be aware of the risks posed by LCMV and should consider the virus in the differential diagnosis of aseptic meningitis, especially in patients who reported contact with rodents. Furthermore, LCMV should be considered in infants and children with unexplained hydrocephalus, intracerebral calcifications and chorioretinitis. Despite intensive interdisciplinary research efforts, efficient antiviral therapy for LCMV infection is still not available.
Collapse
Affiliation(s)
- Tatjana Vilibic-Cavlek
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia; (M.B.); (I.T.); (I.F.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Vladimir Savic
- Laboratory for Virology and Serology, Poultry Center, Croatian Veterinary Institute, 10000 Zagreb, Croatia;
| | - Thomas Ferenc
- Clinical Department of Diagnostic and Interventional Radiology, Merkur University Hospital, 10000 Zagreb, Croatia;
| | - Anna Mrzljak
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
- Department of Gastroenterology and Hepatology, Clinical Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Ljubo Barbic
- Department of Microbiology and Infectious Diseases with Clinic, Faculty of Veterinary Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.B.); (V.S.)
| | - Maja Bogdanic
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia; (M.B.); (I.T.); (I.F.)
| | - Vladimir Stevanovic
- Department of Microbiology and Infectious Diseases with Clinic, Faculty of Veterinary Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.B.); (V.S.)
| | - Irena Tabain
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia; (M.B.); (I.T.); (I.F.)
| | - Ivana Ferencak
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia; (M.B.); (I.T.); (I.F.)
| | - Snjezana Zidovec-Lepej
- Department of Immunological and Molecular Diagnostics, University Hospital for Infectious Diseases “Dr Fran Mihaljevic”, 10000 Zagreb, Croatia;
| |
Collapse
|
31
|
El-Azazy M, Nabil I, Hassan SS, El-Shafie AS. Adsorption Characteristics of Pristine and Magnetic Olive Stones Biochar with Respect to Clofazimine. NANOMATERIALS 2021; 11:nano11040963. [PMID: 33918728 PMCID: PMC8070022 DOI: 10.3390/nano11040963] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/17/2022]
Abstract
Olive stone biochars (OSBC), both pristine and following magnetization (MAG-OSBC), were utilized as eco-friendly and cost-effective sorbents for the antituberculosis, clofazimine (CLOF). Morphologies, textures, surface functionalities, and thermal stabilities of both adsorbents were explored using SEM, EDX, TEM, BET, FT-IR, Raman, XRD and TGA analyses. SEM analysis showed meso- and macroporous surfaces. BET data showed that the MAG-OSBC possesses a larger surface area (33.82 m2/g) and pore volume. Batch adsorption studies were conducted following the experimental scenario of Box-Behnken (BB) design. The adsorption efficiency of both adsorbents was evaluated in terms of the % removal (%R) and the sorption capacity (qe, mg/g). Dependent variables (%R and qe) were maximized as a function of four factors: pH, sorbent dose (AD), the concentration of CLOF ([CLOF]), and contact time (CT). A %R of 98.10% and 98.61% could be obtained using OSBC and MAG-OSBC, respectively. Equilibrium studies indicated that both Langmuir and Freundlich models were perfectly fit for adsorption of CLOF. Maximum adsorption capacity (qmax) of 174.03 mg/g was obtained using MAG-OSBC. Adsorption kinetics could be best illustrated using the pseudo-second-order (PSO) model. The adsorption-desorption studies showed that both adsorbents could be restored with the adsorption efficiency being conserved up to 92% after the sixth cycles.
Collapse
|
32
|
Egiz A, Gala D. Clofazimine: another potential magic bullet for the treatment of COVID-19? Postgrad Med J 2021; 98:e124. [PMID: 33795478 DOI: 10.1136/postgradmedj-2021-140143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2021] [Indexed: 11/04/2022]
Affiliation(s)
- Abdullah Egiz
- School of Medicine, University of Central Lancashire, Preston, UK
| | - Dhir Gala
- School of Medicine, University of Central Lancashire, Preston, UK
| |
Collapse
|
33
|
Herring S, Oda JM, Wagoner J, Kirchmeier D, O'Connor A, Nelson EA, Huang Q, Liang Y, DeWald LE, Johansen LM, Glass PJ, Olinger GG, Ianevski A, Aittokallio T, Paine MF, Fink SL, White JM, Polyak SJ. Inhibition of Arenaviruses by Combinations of Orally Available Approved Drugs. Antimicrob Agents Chemother 2021; 65:e01146-20. [PMID: 33468464 PMCID: PMC8097473 DOI: 10.1128/aac.01146-20] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Neglected diseases caused by arenaviruses such as Lassa virus (LASV) and filoviruses like Ebola virus (EBOV) primarily afflict resource-limited countries, where antiviral drug development is often minimal. Previous studies have shown that many approved drugs developed for other clinical indications inhibit EBOV and LASV and that combinations of these drugs provide synergistic suppression of EBOV, often by blocking discrete steps in virus entry. We hypothesize that repurposing of combinations of orally administered approved drugs provides effective suppression of arenaviruses. In this report, we demonstrate that arbidol, an approved influenza antiviral previously shown to inhibit EBOV, LASV, and many other viruses, inhibits murine leukemia virus (MLV) reporter viruses pseudotyped with the fusion glycoproteins (GPs) of other arenaviruses (Junin virus [JUNV], lymphocytic choriomeningitis virus [LCMV], and Pichinde virus [PICV]). Arbidol and other approved drugs, including aripiprazole, amodiaquine, sertraline, and niclosamide, also inhibit infection of cells by infectious PICV, and arbidol, sertraline, and niclosamide inhibit infectious LASV. Combining arbidol with aripiprazole or sertraline results in the synergistic suppression of LASV and JUNV GP-bearing pseudoviruses. This proof-of-concept study shows that arenavirus infection in vitro can be synergistically inhibited by combinations of approved drugs. This approach may lead to a proactive strategy with which to prepare for and control known and new arenavirus outbreaks.
Collapse
Affiliation(s)
- Shawn Herring
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Jessica M Oda
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Jessica Wagoner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Delaney Kirchmeier
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Aidan O'Connor
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Elizabeth A Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Qinfeng Huang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - Yuying Liang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - Lisa Evans DeWald
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | | | - Pamela J Glass
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | | | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Oslo Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, Oslo, Norway
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Mary F Paine
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Susan L Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Judith M White
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, USA
| | - Stephen J Polyak
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
34
|
Ateya AM. Clofazimine: A potential therapeutic option for severe COVID-19. Med Hypotheses 2021; 150:110535. [PMID: 33735762 PMCID: PMC7880843 DOI: 10.1016/j.mehy.2021.110535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/24/2022]
Affiliation(s)
- Areej Mohamed Ateya
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Institut für Klinische Pharmakologie und Toxikologie, Charité-Universitätsmedizin, Berlin, Germany.
| |
Collapse
|