1
|
Lotfian S, Soleimani A, Taromchi AH, Sabzehei F, Dinmohammadi H, Nedaei K. The recombinant immunodominant regions 179-344 and 550-670 from SARS-COV2 spike protein can efficiently react with patients' sera. Microb Pathog 2025; 205:107604. [PMID: 40287104 DOI: 10.1016/j.micpath.2025.107604] [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: 01/28/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
SARS-COV2 is related to the COVID-19 pandemic that emerged in 2019. It caused severe criticism of world health care and the economic system. Its high mutation rate increases contagiousness and gives rise to new variants, causing recurrent epidemic waves. One of the effective strategies for prevention and controlling infections is evaluating antibody existence against SARS-CoV2 to know how much individuals are protected against new infection. Therefore, it can be helpful to conduct health care strategies for vaccination and healing infected patients. In this study, we identified two immunodominant linear B cell epitopes (S179-344 and S550-670) within the S1 subunit of the spike protein using bioinformatics tools (ABCpred and BepiPred). These regions were subsequently cloned into the pET28a(+) vector, expressed in E. coli BL21 (DE3), and then purified. The recombinant antigens were then assessed for their seroreactivity using a variety of diagnostic techniques, including Western blot, dot blot, and ELISA. The study population for this assessment included 68 positive sera from patients infected with SARS-CoV2 and 63 negative control samples, both of which were screened by standard PCR. The results obtained from the ELISA assay indicated that both antigens demonstrated strong seroreactivity with high sensitivity (91-98 %) and specificity (82 %), suggesting their potential as diagnostic tools. The combination of both antigens exhibited diagnostic accuracy, further supporting their utility in clinical settings. The results of this study underscore the remarkable antigenicity of these epitopes and underscore the suitability of the assay for evaluating community immunity levels against SARS-CoV2 and vaccine efficacy.
Collapse
Affiliation(s)
- Sama Lotfian
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Akbar Soleimani
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Amir Hossein Taromchi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Faezeh Sabzehei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Dinmohammadi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Keivan Nedaei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
2
|
Kılıç G, Demirkan E, Yücel F. Development of Anti-idiotypic Monoclonal Antibody Mimicking SARS-CoV-2 Receptor Binding Domain. Mol Biotechnol 2025; 67:1556-1564. [PMID: 38662257 PMCID: PMC11928402 DOI: 10.1007/s12033-024-01138-1] [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: 10/20/2023] [Accepted: 03/11/2024] [Indexed: 04/26/2024]
Abstract
Using the hybridoma technique, we developed a panel of anti-idiotypic monoclonal antibodies (aId-mAb) that mimic The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Receptor-Binding Domain (RBD) molecule against Fragment antigen-binding (Fab) of anti-SARS-CoV-2 (S1, RBD) antibodies. Investigated the in vivo and in vitro effects of these aId-mAbs we developed and examined their antigenic mimicry abilities. Among these 12 antibodies, 6 aId-mAbs (designated FY1B4, FY2A6, H9F3, E6G7, FY7E11, and FY8H3) were selected for further characterization in a series of experiments. First, competitive receptor binding assay results confirmed that six aId-mAbs could specifically bind to the ACE2 receptor in target cells and block the interaction between the RBD molecule and the ACE receptor. Moreover, we examined the immunological activities of these aId-mAbs in female BALB/c and showed that E6G7, H7E11, and H8H3 aId-mAbs induce an antibody response by mimicking RBD and stimulating the immune system. It is considered that these three aId-mAbs will be evaluated as SARS-CoV-2 vaccine candidate molecules in future studies.
Collapse
Affiliation(s)
- Gamze Kılıç
- Bursa Uludag University, Faculty of Arts and Sciences, Biology Department, Görükle Campus, Bursa, Turkey
- TUBITAK, Marmara Research Center, Life Sciences, Genetic Engineering and Biotechnology, Kocaeli, Turkey
| | - Elif Demirkan
- Bursa Uludag University, Faculty of Arts and Sciences, Biology Department, Görükle Campus, Bursa, Turkey
| | - Fatıma Yücel
- TUBITAK, Marmara Research Center, Life Sciences, Genetic Engineering and Biotechnology, Kocaeli, Turkey.
| |
Collapse
|
3
|
Zheng H, Qiu C, Tian H, Zhu X, Yin B, Zhou Z, Li X, Zhao J. Host restriction factors against porcine epidemic diarrhea virus: a mini-review. Vet Res 2025; 56:67. [PMID: 40128890 PMCID: PMC11934732 DOI: 10.1186/s13567-025-01500-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/31/2024] [Indexed: 03/26/2025] Open
Abstract
Porcine epidemic diarrhea is an acute contagious disease caused by porcine epidemic diarrhea virus (PEDV), which severely constrains the development of the global swine industry. Host restriction factors constitute a vital defensive barrier against viral infections, typically interacting with viruses at specific stages of their replication process to disrupt it. Considering that traditional PEDV vaccines often struggle to effectively activate mucosal immunity in sows and thereby fail to provide reliable passive immunity to piglets via milk, this review focuses on the host restriction factors that play crucial roles in restricting PEDV infection and replication. The aim is to identify potential targets for the development of anti-PEDV drugs and offer insights for the exploration of novel vaccine adjuvants.
Collapse
Affiliation(s)
| | - Cunyi Qiu
- Gansu Polytechnic College of Animal Husbandry & Engineering, Wuwei, 733006, China
| | - Haolun Tian
- Northwest a&F University, Yangling, 712000, China
| | - Xiaofu Zhu
- Xianyang Polytechnic Institute, Xianyang, 712000, China
| | - Baoying Yin
- Xianyang Polytechnic Institute, Xianyang, 712000, China
| | - Zhiding Zhou
- Key Laboratory of Marine Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xuezhao Li
- Gansu Polytechnic College of Animal Husbandry & Engineering, Wuwei, 733006, China
| | - Jingjing Zhao
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
4
|
Xiao W, Li Z, Chen C, Shi Y, Fang P, Xiao S, Fang L. Revisiting the roles of trypsin in the productive infection of porcine deltacoronavirus in porcine-derived cells. Virology 2025; 604:110453. [PMID: 39961260 DOI: 10.1016/j.virol.2025.110453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/25/2025] [Accepted: 02/12/2025] [Indexed: 05/09/2025]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging enteric coronavirus with the potential for interspecies transmission. Trypsin has been shown to play a positive role in the isolation and multiplication of PDCoV in vitro, however, the functions of trypsin during PDCoV replication cycle remain controversial. In this study, we revisited the roles of trypsin for PDCoV infection by utilizing two kinds of PDCoV, PDCoVT+ and PDCoVT-, which were prepared in the presence or absence of trypsin, respectively. We found that PDCoVT+ was able to continuously proliferate in the medium containing trypsin, achieving a higher titer as the infection progress in LLC-PK1 and other tested porcine-derived cells. However, its replication was only transiently improved at 12 hours post-infection, and lower viral titers were observed under trypsin-free culture conditions. Furthermore, the trypsin-mediated enhancement of viral replication could be inhibited by trypsin inhibitor SBTI, suggesting that the second-round viral reproduction of PDCoVT+ might be impeded without trypsin. We further investigated the replication dynamics of PDCoVT- in LLC-PK1 cells in the presence or absence of trypsin. The results indicated that PDCoVT- generated lower viral titers under trypsin-free culture conditions, while the addition of trypsin reverted the infectivity of PDCoVT-. Additionally, we demonstrated that trypsin cleaved the PDCoV spike protein, activating viral attachment and internalization. Moreover, trypsin promoted viral replication and release, accelerating PDCoV maturation and facilitating second-round infection. Taken together, this study systematically revaluated and emphasized an essential role of trypsin in PDCoV infection, providing mechanistic insights into the productive infection of PDCoV in porcine-derived cells.
Collapse
Affiliation(s)
- Wenwen Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Zhuang Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Chaoqun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuting Shi
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Puxian Fang
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Liurong Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
5
|
Del Sorbo L, Giugliano R, Cerracchio C, Iovane V, Salvatore MM, Serra F, Amoroso MG, Pellegrini F, Levante M, Capozza P, Diakoudi G, Galdiero M, Fusco G, Pratelli A, Andolfi A, Fiorito F. In Vitro Evaluation of Aryl Hydrocarbon Receptor Involvement in Feline Coronavirus Infection. Viruses 2025; 17:227. [PMID: 40006982 PMCID: PMC11860311 DOI: 10.3390/v17020227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Feline coronavirus (FCoV) is an alphacoronavirus (αCoV) that causes moderate or chronic asymptomatic infection in cats. However, in a single infected cat, FCoV can modify its cellular tropism by acquiring the ability to infect macrophages, resulting in the development of feline infectious peritonitis (FIP). In this context, to restrain the impact of FCoV infection, scientific research has focused attention on the development of antiviral therapies involving novel mechanisms of action. Recent studies have demonstrated that aryl hydrocarbon receptor (AhR) signaling regulates the host response to different human and animal CoVs. Hence, the mechanism of action of AhR was evaluated upon FCoV infection in Crandell Feline Kidney (CRFK) and in canine fibrosarcoma (A72) cells. Following infection with feline enteric CoV (FECV), strain "München", a significant activation of AhR and of its target CYP1A1, was observed. The selective AhR antagonist CH223191 provoked a reduction in FCoV replication and in the levels of viral nucleocapsid protein (NP). Furthermore, the effect of the AhR inhibitor on the acidity of lysosomes in infected cells was observed. Our findings indicate that FCoV acts on viral replication that upregulates AhR. CH223191 repressed virus yield through the inhibition of AhR. In this respect, for counteracting FCoV, AhR represents a new target useful for identifying antiviral drugs. Moreover, in the presence of CH223191, the alkalinization of lysosomes in FCoV-infected CRFK cells was detected, outlining their involvement in antiviral activity.
Collapse
Affiliation(s)
- Luca Del Sorbo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
| | - Rosa Giugliano
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Claudia Cerracchio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
| | - Valentina Iovane
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy; (V.I.); (A.A.)
| | - Maria Michela Salvatore
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Francesco Serra
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Maria Grazia Amoroso
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Francesco Pellegrini
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Martina Levante
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Paolo Capozza
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Georgia Diakoudi
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Giovanna Fusco
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Annamaria Pratelli
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Anna Andolfi
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy; (V.I.); (A.A.)
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, Italy
| | - Filomena Fiorito
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, Italy
| |
Collapse
|
6
|
Mao B, Le-Trilling VTK, Tang H, Hu J, Schmitz MS, Barbet K, Xu D, Wei Z, Guo B, Mennerich D, Yao C, Liu J, Li Z, Wan Y, Zhang X, Wang K, Tang N, Yu Z, Trilling M, Lin Y. Diphyllin elicits a doubled-pronged attack on the entry of SARS-CoV-2 by inhibiting cathepsin L and furin. Virus Res 2024; 350:199485. [PMID: 39424146 PMCID: PMC11532987 DOI: 10.1016/j.virusres.2024.199485] [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: 12/28/2023] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the coronavirus disease 2019 (COVID-19) pandemic, posing serious threats to global health. Effective broad-spectrum antiviral drugs for the treatment of COVID-19 are not sufficiently available. In the present study, we investigated the antiviral activity of the natural lignan diphyllin (PubChem CID 100492) against different SARS-CoV-2 variants and explored the underlying molecular mechanisms. We found that diphyllin dose-dependently inhibits the SARS-CoV-2 spike (S)-mediated entry into different types of cells. The potent inhibition was evident against spike proteins derived from the original SARS-CoV-2 and from variants of concern such as Alpha, Beta, Delta or Omicron. Accordingly, diphyllin also significantly inhibited the in vitro infection of a clinical SARS-CoV-2 virus isolate. Mechanistically, diphyllin simultaneously inhibited the endosomal entry of SARS-CoV-2 by neutralizing the endosomal acidification and reducing the activity of the cysteine protease cathepsin L (CTSL) as well as S-meditated cell surface entry by impairing furin activity. Collectively, our findings establish diphyllin as novel inhibitor of CTSL and furin proteases, resulting in a double-pronged attack on SARS-CoV-2 entry along endosomal as well as cell surface routes. Therefore, diphyllin has the potential to be advanced as an inhibitor of SARS-CoV-2 entry.
Collapse
Affiliation(s)
- Binli Mao
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China; Department of Blood Transfusion, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Vu Thuy Khanh Le-Trilling
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany; Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Haihuan Tang
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Jie Hu
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China; Department of Laboratory Medicine, Bishan Hospital of Chongqing Medical University, Chongqing 402760, China
| | - Mona S Schmitz
- Department of Pulmonary Medicine, University Medical Center Essen, Ruhrlandklinik, Essen 45239, Germany
| | - Kimberly Barbet
- Department of Pulmonary Medicine, University Medical Center Essen, Ruhrlandklinik, Essen 45239, Germany
| | - Dan Xu
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Zhen Wei
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Beinu Guo
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Denise Mennerich
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Chun Yao
- Chongqing Yucai Secondary School, Chongqing 400050, China
| | - Jinxin Liu
- Chongqing Yucai Secondary School, Chongqing 400050, China
| | - Zhenghan Li
- Chongqing Yucai Secondary School, Chongqing 400050, China
| | - Yushun Wan
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoyong Zhang
- Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kai Wang
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Ni Tang
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Zebo Yu
- Department of Blood Transfusion, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Mirko Trilling
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany; Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany.
| | - Yong Lin
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
7
|
Schwerdtner M, Schmacke LC, Nave J, Limburg H, Steinmetzer T, Stein DA, Moulton HM, Böttcher-Friebertshäuser E. Unveiling the Role of TMPRSS2 in the Proteolytic Activation of Pandemic and Zoonotic Influenza Viruses and Coronaviruses in Human Airway Cells. Viruses 2024; 16:1798. [PMID: 39599912 PMCID: PMC11599139 DOI: 10.3390/v16111798] [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: 10/16/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
The zoonotic transmission of influenza A viruses (IAVs) and coronaviruses (CoVs) may result in severe disease. Cleavage of the surface glycoproteins hemagglutinin (HA) and spike protein (S), respectively, is essential for viral infectivity. The transmembrane serine protease 2 (TMPRSS2) is crucial for cleaving IAV HAs containing monobasic cleavage sites and severe acute respiratory syndrome (SARS)-CoV-2 S in human airway cells. Here, we analysed and compared the TMPRSS2-dependency of SARS-CoV, Middle East respiratory syndrome (MERS)-CoV, the 1918 pandemic H1N1 IAV and IAV H12, H13 and H17 subtypes in human airway cells. We used the peptide-conjugated morpholino oligomer (PPMO) T-ex5 to knockdown the expression of active TMPRSS2 and determine the impact on virus activation and replication in Calu-3 cells. The activation of H1N1/1918 and H13 relied on TMPRSS2, whereas recombinant IAVs carrying H12 or H17 were not affected by TMPRSS2 knockdown. MERS-CoV replication was strongly suppressed in T-ex5 treated cells, while SARS-CoV was less dependent on TMPRSS2. Our data underline the importance of TMPRSS2 for certain (potentially) pandemic respiratory viruses, including H1N1/1918 and MERS-CoV, in human airways, further suggesting a promising drug target. However, our findings also highlight that IAVs and CoVs differ in TMPRSS2 dependency and that other proteases are involved in virus activation.
Collapse
Affiliation(s)
- Marie Schwerdtner
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany; (M.S.)
| | - Luna C. Schmacke
- Institute of Pharmaceutical Chemistry, Philipps-University Marburg, 35037 Marburg, Germany
| | - Julia Nave
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany; (M.S.)
| | - Hannah Limburg
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany; (M.S.)
| | - Torsten Steinmetzer
- Institute of Pharmaceutical Chemistry, Philipps-University Marburg, 35037 Marburg, Germany
| | - David A. Stein
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Hong M. Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | | |
Collapse
|
8
|
Hoffmann M, Kleine-Weber H, Graichen L, Nehlmeier I, Kempf A, Moldenhauer AS, Braun E, Assiri AM, Kirchhoff F, Sauter D, Alkharsah KR, Pöhlmann S. Acquisition of a multibasic cleavage site does not increase MERS-CoV entry into Calu-3 human lung cells. J Virol 2024; 98:e0130524. [PMID: 39470207 PMCID: PMC11575293 DOI: 10.1128/jvi.01305-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024] Open
Abstract
Human-to-human transmission of the highly pathogenic Middle East respiratory syndrome coronavirus (MERS-CoV) is currently inefficient. However, there is concern that the virus might mutate and thereby increase its transmissibility and thus pandemic potential. The pandemic SARS-CoV-2 depends on a highly cleavable furin motif at the S1/S2 site of the viral spike (S) protein for efficient lung cell entry, transmission, and pathogenicity. Here, by employing pseudotyped particles, we investigated whether augmented cleavage at the S1/S2 site also increases MERS-CoV entry into Calu-3 human lung cells. We report that polymorphism T746K at the S1/S2 cleavage site or optimization of the furin motif increases S protein cleavage but not lung cell entry. These findings suggest that, unlike what has been reported for SARS-CoV-2, a highly cleavable S1/S2 site might not augment MERS-CoV infectivity for human lung cells.IMPORTANCEThe highly cleavable furin motif in the spike protein is required for robust lung cell entry, transmission, and pathogenicity of SARS-CoV-2. In contrast, it is unknown whether optimization of the furin motif in the spike protein of the pre-pandemic MERS-CoV increases lung cell entry and allows for robust human-human transmission. The present study indicates that this might not be the case. Thus, neither a naturally occurring polymorphism that increased MERS-CoV spike protein cleavage nor artificial optimization of the cleavage site allowed for increased spike-protein-driven entry into Calu-3 human lung cells.
Collapse
Affiliation(s)
- Markus Hoffmann
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Hannah Kleine-Weber
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Luise Graichen
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Anna-Sophie Moldenhauer
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
| | - Elisabeth Braun
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Daniel Sauter
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Khaled R. Alkharsah
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University (IAU), Dammam, Saudi Arabia
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| |
Collapse
|
9
|
Dey S, Pahari P, Mukherjee S, Munro JB, Das DK. Conformational dynamics of SARS-CoV-2 Omicron spike trimers during fusion activation at single molecule resolution. Structure 2024; 32:1910-1925.e6. [PMID: 39366371 PMCID: PMC11560620 DOI: 10.1016/j.str.2024.09.008] [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: 05/06/2024] [Revised: 07/26/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron entry involves spike (S) glycoprotein-mediated fusion of viral and late endosomal membranes. Here, using single-molecule Förster resonance energy transfer (sm-FRET) imaging and biochemical measurements, we directly visualized conformational changes of individual spike trimers on the surface of SARS-CoV-2 Omicron pseudovirions during fusion activation. We observed that the S2 domain of the Omicron spike is a dynamic fusion machine. S2 reversibly interchanges between the pre-fusion conformation and two previously undescribed intermediate conformations. Acidic pH shifts the conformational equilibrium of S2 toward an intermediate conformation and promotes the membrane hemi-fusion reaction. Moreover, we captured conformational reversibility in the S2 domain, which suggests that spike can protect itself from pre-triggering. Furthermore, we determined that Ca2+ directly promotes the S2 conformational change from an intermediate conformation to post-fusion conformation. In the presence of a target membrane, low pH and Ca2+ stimulate the irreversible transition to S2 post-fusion state and promote membrane fusion.
Collapse
Affiliation(s)
- Shuvankar Dey
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Purba Pahari
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Srija Mukherjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - James B Munro
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dibyendu Kumar Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India; Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.
| |
Collapse
|
10
|
Song C, Li H, Han Y, Wang K, Yan W, Yang X, Zhang A, Wang H. Host restriction factor Rab11a limits Porcine deltacoronavirus invasion of cells via fusion peptide-mediated membrane fusion. Vet Microbiol 2024; 298:110246. [PMID: 39244909 DOI: 10.1016/j.vetmic.2024.110246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
Porcine deltacoronavirus (PDCoV) poses a serious threat to pork industry and has the potential for cross-species transmission. Yet, the invasion mechanisms and host factors involved are still unknown. In the present work, using siRNA interference and co-immunoprecipitation, we identified Annexin A2 (ANXA2), Prohibitin-2 (PHB2), or Caveolin-2 (CAV2) as host factors positively regulating the internalization of PDCoV. We further found that Rab11a co-localized with PDCoV S and inhibited PDCoV internalization. Subsequently, a pseudoviral infection model (LV-PDCoV S-GFP) was constructed, and ANXA2 or CAV2 promoted PDCoV invasion by downregulating Rab11a. Our results also indicated that ANXA2, CAV2, and Rab11a interact with the S protein via S-FP, thereby regulating virus-host membrane fusion. Through LV-PDCoV S-GFP infection, we found that Rab11a may act as a host restriction factor, and it could regulate the invasion efficiency of PDCoV by adding of exogenous GTP. These findings revealed that Rab11a was an exciting target to restrict fusion of PDCoV with host cell membranes. AVAILABILITY OF DATA AND MATERIAL: Not applicable.
Collapse
Affiliation(s)
- Cailiang Song
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Hao Li
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yun Han
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Kailu Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Wenjun Yan
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Xin Yang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| | - Anyun Zhang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| | - Hongning Wang
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610000, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| |
Collapse
|
11
|
Singh P, Pahari P, Mukherjee S, Karmakar S, Hoffmann M, Mandal T, Das DK. SARS-CoV-2 spike fusion peptide trans interaction with phosphatidylserine lipid triggers membrane fusion for viral entry. mBio 2024; 15:e0107724. [PMID: 39115315 PMCID: PMC11389415 DOI: 10.1128/mbio.01077-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/30/2024] [Indexed: 09/12/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike is the fusion machine for host cell entry. Still, the mechanism by which spike protein interacts with the target lipid membrane to facilitate membrane fusion during entry is not fully understood. Here, using steady-state membrane fusion and single-molecule fluorescence resonance energy transfer imaging of spike trimers on the surface of SARS-CoV-2 pseudovirion, we directly show that spike protein interacts with phosphatidylserine (PS) lipid in the target membrane for mediating fusion. We observed that the fusion peptide of the spike S2 domain interacts with the PS lipid of the target membrane. Low pH and Ca2+ trigger the spike conformational change and bring fusion peptide in close proximity to the PS lipid of the membrane. The binding of the spike with PS lipid of its viral membrane (cis interaction) impedes the fusion activation. PS on the target membrane promotes spike binding via trans interaction, prevents the cis interaction, and accelerates fusion. Sequestering or absence of PS lipid abrogates the spike-mediated fusion process and restricts SARS-CoV-2 infectivity. We found that PS-dependent interaction for fusion is conserved across all the SARS-CoV-2 spike variants of concern (D614G, Alpha, Beta, Delta, and Omicron). Our study suggests that PS lipid is indispensable for SARS-CoV-2 spike-mediated virus and target membrane fusion for entry, and restricting PS interaction with spike inhibits the SARS-CoV-2 spike-mediated entry. Therefore, PS is an important cofactor and acts as a molecular beacon in the target membrane for SARS-CoV-2 entry. IMPORTANCE The role of lipids in the host cell target membrane for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry is not clear. We do not know whether SARS-CoV-2 spike protein has any specificity in terms of lipid for membrane fusion reaction. Here, using in vitro reconstitution of membrane fusion assay and single-molecule fluorescence resonance energy transfer imaging of SARS-CoV-2 spike trimers on the surface of the virion, we have demonstrated that phosphatidylserine (PS) lipid plays a key role in SARS-CoV-2 spike-mediated membrane fusion reaction for entry. Membrane-externalized PS lipid strongly promotes spike-mediated membrane fusion and COVID-19 infection. Blocking externalized PS lipid with PS-binding protein or in the absence of PS, SARS-CoV-2 spike-mediated fusion is strongly inhibited. Therefore, PS is an important target for restricting viral entry and intervening spike, and PS interaction presents new targets for COVID-19 interventions.
Collapse
Affiliation(s)
- Puspangana Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Purba Pahari
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Srija Mukherjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Sharmistha Karmakar
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland, USA
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center—Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg August University, Göttingen, Germany
| | - Taraknath Mandal
- Department of Physics, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Dibyendu Kumar Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
- Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| |
Collapse
|
12
|
Maleš P, Nikšić-Franjić I, Wang A, Pem B, Bakarić D. Optical and molecular features of negatively curved surfaces created by POPE lipids: A crucial role of the initial conditions. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 317:124462. [PMID: 38754204 DOI: 10.1016/j.saa.2024.124462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/18/2024]
Abstract
Membrane fusion is closely related to plasma membrane domains rich in cone-shaped phosphatidylethanolamine (PE) lipids that can reverse membrane curvature under certain conditions. The phase transition of PE-based lipid membranes from the lamellar fluid phase (Lα) to the inverse hexagonal phase (HII) is commonly taken as a general model in reconstructing the membrane fusion pathway, and whose structural features have been mostly described so far using structural and microscopic techniques. The aim of this paper is to decipher the optical and molecular features of Lβ → Lα and especially of Lα → HII transition of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE) lipids at pH = 7.0 when they are initially prepared in the form of both multi- and unilamellar liposomes (MLVs and LUVs). The distinction between optical properties of MLS- and LUVs-derived HII phase, provided from turbidity-sensitive temperature-dependent UV-Vis spectra, was attributed to different formation mechanisms of HII phase. Most importantly, from FTIR spectroscopic data of POPE lipids in Lβ (15 °C), Lα (50 °C) and HII (85 °C) phases we identified the changes in molecular features of POPE lipids during phase transitions. Among the latter, by far the most significant is different hydration pattern of POPE lipids in MLVs- and LUVs-derived HII phase which extends from the polar-apolar interface all the way to the terminal amino group of the POPE lipid, along with the changes in the conformation of glycerol backbone as evidenced by the signature of α-methylene groups. Molecular dynamics simulations confirmed higher water penetration in HII phase and provided insight into hydrogen bonding patterns.
Collapse
Affiliation(s)
- Petra Maleš
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia
| | - Ivana Nikšić-Franjić
- School of Science, Constructor University, Bremen GmbH, Campus Ring 1, 28759 Bremen, Germany
| | - Anna Wang
- School of Chemistry, Australian Centre for Astrobiology, and ARC Centre of Excellence in Synthetic Biology, University of New South Wales Sydney, Bedegal Country, Sydney, NSW 2052, Australia
| | - Barbara Pem
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia
| | - Danijela Bakarić
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia.
| |
Collapse
|
13
|
Bolsinger MM, Drobny A, Wilfling S, Reischl S, Krach F, Moritz R, Balta D, Hehr U, Sock E, Bleibaum F, Hanses F, Winner B, Huarcaya SP, Arnold P, Zunke F. SARS-CoV-2 Spike Protein Induces Time-Dependent CTSL Upregulation in HeLa Cells and Alveolarspheres. J Cell Biochem 2024; 125:e30627. [PMID: 38971996 DOI: 10.1002/jcb.30627] [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: 03/27/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/08/2024]
Abstract
Autophagy and lysosomal pathways are involved in the cell entry of SARS-CoV-2 virus. To infect the host cell, the spike protein of SARS-CoV-2 binds to the cell surface receptor angiotensin-converting enzyme 2 (ACE2). To allow the fusion of the viral envelope with the host cell membrane, the spike protein has to be cleaved. One possible mechanism is the endocytosis of the SARS-CoV-2-ACE2 complex and subsequent cleavage of the spike protein, mainly by the lysosomal protease cathepsin L. However, detailed molecular and dynamic insights into the role of cathepsin L in viral cell entry remain elusive. To address this, HeLa cells and iPSC-derived alveolarspheres were treated with recombinant SARS-CoV-2 spike protein, and the changes in mRNA and protein levels of cathepsins L, B, and D were monitored. Additionally, we studied the effect of cathepsin L deficiency on spike protein internalization and investigated the influence of the spike protein on cathepsin L promoters in vitro. Furthermore, we analyzed variants in the genes coding for cathepsin L, B, D, and ACE2 possibly associated with disease progression using data from Regeneron's COVID Results Browser and our own cohort of 173 patients with COVID-19, exhibiting a variant of ACE2 showing significant association with COVID-19 disease progression. Our in vitro studies revealed a significant increase in cathepsin L mRNA and protein levels following exposure to the SARS-CoV-2 spike protein in HeLa cells, accompanied by elevated mRNA levels of cathepsin B and D in alveolarspheres. Moreover, an increase in cathepsin L promoter activity was detected in vitro upon spike protein treatment. Notably, the knockout of cathepsin L resulted in reduced internalization of the spike protein. The study highlights the importance of cathepsin L and lysosomal proteases in the SARS-CoV-2 spike protein internalization and suggests the potential of lysosomal proteases as possible therapeutic targets against COVID-19 and other viral infections.
Collapse
Affiliation(s)
- Magdalena M Bolsinger
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Stephanie Reischl
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Krach
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raul Moritz
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Denise Balta
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ute Hehr
- Center for Human Genetics Regensburg, Regensburg, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Bleibaum
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Frank Hanses
- Emergency Department, University Hospital Regensburg, Regensburg, Germany
- Department for Infection Control and Infectious Diseases, University Hospital Regensburg, Regensburg, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Susy Prieto Huarcaya
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Philipp Arnold
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
14
|
Dai X, Xu R, Li N. The Interplay between Airway Cilia and Coronavirus Infection, Implications for Prevention and Control of Airway Viral Infections. Cells 2024; 13:1353. [PMID: 39195243 PMCID: PMC11353096 DOI: 10.3390/cells13161353] [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: 07/23/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Coronaviruses (CoVs) are a class of respiratory viruses with the potential to cause severe respiratory diseases by infecting cells of the upper respiratory tract, bronchial epithelium, and lung. The airway cilia are distributed on the surface of respiratory epithelial cells, forming the first point of contact between the host and the inhaled coronaviruses. The function of the airway cilia is to oscillate and sense, thereby defending against and removing pathogens to maintain the cleanliness and patency of the respiratory tract. Following infection of the respiratory tract, coronaviruses exploit the cilia to invade and replicate in epithelial cells while also damaging the cilia to facilitate the spread and exacerbation of respiratory diseases. It is therefore imperative to investigate the interactions between coronaviruses and respiratory cilia, as well as to elucidate the functional mechanism of respiratory cilia following coronavirus invasion, in order to develop effective strategies for the prevention and treatment of respiratory viral infections. This review commences with an overview of the fundamental characteristics of airway cilia, and then, based on the interplay between airway cilia and coronavirus infection, we propose that ciliary protection and restoration may represent potential therapeutic approaches in emerging and re-emerging coronavirus pandemics.
Collapse
Affiliation(s)
| | - Ruodan Xu
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| | - Ning Li
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| |
Collapse
|
15
|
Michaels TM, Essop MF, Joseph DE. Potential Effects of Hyperglycemia on SARS-CoV-2 Entry Mechanisms in Pancreatic Beta Cells. Viruses 2024; 16:1243. [PMID: 39205219 PMCID: PMC11358987 DOI: 10.3390/v16081243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The COVID-19 pandemic has revealed a bidirectional relationship between SARS-CoV-2 infection and diabetes mellitus. Existing evidence strongly suggests hyperglycemia as an independent risk factor for severe COVID-19, resulting in increased morbidity and mortality. Conversely, recent studies have reported new-onset diabetes following SARS-CoV-2 infection, hinting at a potential direct viral attack on pancreatic beta cells. In this review, we explore how hyperglycemia, a hallmark of diabetes, might influence SARS-CoV-2 entry and accessory proteins in pancreatic β-cells. We examine how the virus may enter and manipulate such cells, focusing on the role of the spike protein and its interaction with host receptors. Additionally, we analyze potential effects on endosomal processing and accessory proteins involved in viral infection. Our analysis suggests a complex interplay between hyperglycemia and SARS-CoV-2 in pancreatic β-cells. Understanding these mechanisms may help unlock urgent therapeutic strategies to mitigate the detrimental effects of COVID-19 in diabetic patients and unveil if the virus itself can trigger diabetes onset.
Collapse
Affiliation(s)
- Tara M. Michaels
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| | - M. Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa;
| | - Danzil E. Joseph
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| |
Collapse
|
16
|
Abisheva S, Rutskaya-Moroshan K, Nuranova G, Batyrkhan T, Abisheva A. Antimalarial Drugs at the Intersection of SARS-CoV-2 and Rheumatic Diseases: What Are the Potential Opportunities? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1171. [PMID: 39064600 PMCID: PMC11279047 DOI: 10.3390/medicina60071171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: The coronavirus disease of 2019 (COVID-19) pandemic has posed a serious threat to humanity and is considered a global health emergency. Antimalarial drugs (ADs) have been used in the treatment of immuno-inflammatory arthritis (IIA) and coronavirus infection (COVID-19). The aim of this review is to analyze the current knowledge about the immunomodulatory and antiviral mechanisms of action, characteristics of use, and side effects of antimalarial drugs. Material and Methods: A literature search was carried out using PubMed, MEDLINE, SCOPUS, and Google Scholar databases. The inclusion criteria were the results of randomized and cohort studies, meta-analyses, systematic reviews, and original full-text manuscripts in the English language containing statistically confirmed conclusions. The exclusion criteria were summary reports, newspaper articles, and personal messages. Qualitative methods were used for theoretical knowledge on antimalarial drug usage in AIRDs and SARS-CoV-2 such as a summarization of the literature and a comparison of the treatment methods. Results: The ADs were considered a "candidate" for the therapy of a new coronavirus infection due to mechanisms of antiviral activity, such as interactions with endocytic pathways, the prevention of glycosylation of the ACE2 receptors, blocking sialic acid receptors, and reducing the manifestations of cytokine storms. The majority of clinical trials suggest no role of antimalarial drugs in COVID-19 treatment or prevention. These circumstances do not allow for their use in the treatment and prevention of COVID-19. Conclusions: The mechanisms of hydroxychloroquine are related to potential cardiotoxic manifestations and demonstrate potential adverse effects when used for COVID-19. Furthermore, the need for high doses in the treatment of viral infections increases the likelihood of gastrointestinal side effects, the prolongation of QT, and retinopathy. Large randomized clinical trials (RCTs) have refuted the fact that there is a positive effect on the course and results of COVID-19.
Collapse
Affiliation(s)
- Saule Abisheva
- Department of Family Medicine №1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan; (S.A.); (T.B.); (A.A.)
| | - Kristina Rutskaya-Moroshan
- Department of Family Medicine №1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan; (S.A.); (T.B.); (A.A.)
| | - Gulnaz Nuranova
- Department of Children’s Diseases with Courses in Pulmonology and Nephrology, NJSC “Astana Medical University”, Astana 010000, Kazakhstan;
| | - Tansholpan Batyrkhan
- Department of Family Medicine №1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan; (S.A.); (T.B.); (A.A.)
| | - Anilim Abisheva
- Department of Family Medicine №1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan; (S.A.); (T.B.); (A.A.)
| |
Collapse
|
17
|
Steiner S, Kratzel A, Barut GT, Lang RM, Aguiar Moreira E, Thomann L, Kelly JN, Thiel V. SARS-CoV-2 biology and host interactions. Nat Rev Microbiol 2024; 22:206-225. [PMID: 38225365 DOI: 10.1038/s41579-023-01003-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 01/17/2024]
Abstract
The zoonotic emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing coronavirus disease 2019 (COVID-19) pandemic have profoundly affected our society. The rapid spread and continuous evolution of new SARS-CoV-2 variants continue to threaten global public health. Recent scientific advances have dissected many of the molecular and cellular mechanisms involved in coronavirus infections, and large-scale screens have uncovered novel host-cell factors that are vitally important for the virus life cycle. In this Review, we provide an updated summary of the SARS-CoV-2 life cycle, gene function and virus-host interactions, including recent landmark findings on general aspects of coronavirus biology and newly discovered host factors necessary for virus replication.
Collapse
Affiliation(s)
- Silvio Steiner
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Annika Kratzel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - G Tuba Barut
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto M Lang
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Etori Aguiar Moreira
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Lisa Thomann
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jenna N Kelly
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
- European Virus Bioinformatics Center, Jena, Germany
| | - Volker Thiel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland.
- European Virus Bioinformatics Center, Jena, Germany.
| |
Collapse
|
18
|
Matveev EV, Ponomarev GV, Kazanov MD. Genome-wide bioinformatics analysis of human protease capacity for proteolytic cleavage of the SARS-CoV-2 spike glycoprotein. Microbiol Spectr 2024; 12:e0353023. [PMID: 38189333 PMCID: PMC10846095 DOI: 10.1128/spectrum.03530-23] [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: 10/03/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) primarily enters the cell by binding the virus's spike (S) glycoprotein to the angiotensin-converting enzyme 2 receptor on the cell surface, followed by proteolytic cleavage by host proteases. Studies have identified furin and transmembrane protease serine 2 proteases in priming and triggering cleavages of the S glycoprotein, converting it into a fusion-competent form and initiating membrane fusion, respectively. Alternatively, SARS-CoV-2 can enter the cell through the endocytic pathway, where activation is triggered by lysosomal cathepsin L. However, other proteases are also suspected to be involved in both entry routes. In this study, we conducted a genome-wide bioinformatics analysis to explore the capacity of human proteases in hydrolyzing peptide bonds of the S glycoprotein. Predictive models of sequence specificity for 169 human proteases were constructed and applied to the S glycoprotein together with the method for predicting structural susceptibility to proteolysis of protein regions. After validating our approach on extensively studied S2' and S1/S2 cleavage sites, we applied our method to each peptide bond of the S glycoprotein across all 169 proteases. Our results indicate that various members of the proprotein convertase subtilisin/kexin type, type II transmembrane family serine protease, and kallikrein families, as well as specific coagulation factors, are capable of cleaving S2' or S1/S2 sites. We have also identified a potential cleavage site of cathepsin L at the K790 position within the S2' loop. Structural analysis suggests that cleavage of this site induces conformational changes similar to the cleavage at the R815 (S2') position, leading to the exposure of the fusion peptide and subsequent fusion with the membrane. Other potential cleavage sites and the influence of mutations in common SARS-CoV-2 variants on proteolytic efficiency are discussed.IMPORTANCEThe entry of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) into the cell, activated by host proteases, is considerably more complex in coronaviruses than in most other viruses and is not fully understood. There is evidence that other proteases beyond the known furin and transmembrane protease serine 2 can activate the spike protein. Another example of uncertainty is the cleavage site for the alternative endocytic route of SARS-CoV-2 entrance, which is still unknown. Bioinformatics methods, modeling protease specificity and estimating the structural susceptibility of protein regions to proteolysis, can aid in studying this topic by predicting the involved proteases and their cleavage sites, thereby substantially reducing the amount of experimental work. Elucidating the mechanisms of spike protein activation is crucial for preventing possible future coronavirus pandemics and developing antiviral drugs.
Collapse
Affiliation(s)
- Evgenii V. Matveev
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, Russia
- Research and Training Center on Bioinformatics, A.A.Kharkevich Institute for Information Transmission Problems, Moscow, Russia
- Laboratory of Cytogenetics and Molecular Genetics, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Gennady V. Ponomarev
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, Russia
- Research and Training Center on Bioinformatics, A.A.Kharkevich Institute for Information Transmission Problems, Moscow, Russia
| | - Marat D. Kazanov
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, Russia
- Research and Training Center on Bioinformatics, A.A.Kharkevich Institute for Information Transmission Problems, Moscow, Russia
- Laboratory of Cytogenetics and Molecular Genetics, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| |
Collapse
|
19
|
Doijen J, Heo I, Temmerman K, Vermeulen P, Diels A, Jaensch S, Burcin M, Van den Broeck N, Raeymaekers V, Peremans J, Konings K, Clement M, Peeters D, Van Loock M, Koul A, Buyck C, Van Gool M, Van Damme E. A flexible, image-based, high-throughput platform encompassing in-depth cell profiling to identify broad-spectrum coronavirus antivirals with limited off-target effects. Antiviral Res 2024; 222:105789. [PMID: 38158129 DOI: 10.1016/j.antiviral.2023.105789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The recent pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) posed a major threat to global health. Although the World Health Organization ended the public health emergency status, antiviral drugs are needed to address new variants of SARS-CoV-2 and future pandemics. To identify novel broad-spectrum coronavirus drugs, we developed a high-content imaging platform compatible with high-throughput screening. The platform is broadly applicable as it can be adapted to include various cell types, viruses, antibodies, and dyes. We demonstrated that the antiviral activity of compounds against SARS-CoV-2 variants (Omicron BA.5 and Omicron XBB.1.5), SARS-CoV, and human coronavirus 229E could easily be assessed. The inclusion of cellular dyes and immunostaining in combination with in-depth image analysis enabled us to identify compounds that induced undesirable phenotypes in host cells, such as changes in cell morphology or in lysosomal activity. With the platform, we screened ∼900K compounds and triaged hits, thereby identifying potential candidate compounds carrying broad-spectrum activity with limited off-target effects. The flexibility and early-stage identification of compounds with limited host cell effects provided by this high-content imaging platform can facilitate coronavirus drug discovery. We anticipate that its rapid deployability and fast turnaround can also be applied to combat future pandemics.
Collapse
Affiliation(s)
- Jordi Doijen
- Global Public Health R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Inha Heo
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Koen Temmerman
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Peter Vermeulen
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Annick Diels
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Steffen Jaensch
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Mark Burcin
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | | | | | - Joren Peremans
- Charles River Laboratories, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Katrien Konings
- Charles River Laboratories, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Maxime Clement
- Charles River Laboratories, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Danielle Peeters
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Marnix Van Loock
- Global Public Health R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Anil Koul
- Global Public Health R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Christophe Buyck
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Michiel Van Gool
- Therapeutics Discovery R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Ellen Van Damme
- Global Public Health R&D, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.
| |
Collapse
|
20
|
Jahanshahi S, Ouyang H, Ahmed C, Zahedi Amiri A, Dahal S, Mao YQ, Van Ommen DAJ, Malty R, Duan W, Been T, Hernandez J, Mangos M, Nurtanto J, Babu M, Attisano L, Houry WA, Moraes TJ, Cochrane A. Broad spectrum post-entry inhibitors of coronavirus replication: Cardiotonic steroids and monensin. Virology 2024; 589:109915. [PMID: 37931588 DOI: 10.1016/j.virol.2023.109915] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023]
Abstract
A small molecule screen identified several cardiotonic steroids (digitoxin and ouabain) and the ionophore monensin as potent inhibitors of HCoV-229E, HCoV-OC43, and SARS-CoV-2 replication with EC50s in the low nM range. Subsequent tests confirmed antiviral activity in primary cell models including human nasal epithelial cells and lung organoids. Addition of digitoxin, ouabain, or monensin strongly reduced viral gene expression as measured by both viral protein and RNA accumulation. Furthermore, the compounds acted post virus entry. While the antiviral activity of digitoxin was dependent upon activation of the MEK and JNK signaling pathways but not signaling through GPCRs, the antiviral effect of monensin was reversed upon inhibition of several signaling pathways. Together, the data demonstrates the potent anti-coronavirus properties of two classes of FDA approved drugs that function by altering the properties of the infected cell, rendering it unable to support virus replication.
Collapse
Affiliation(s)
- Shahrzad Jahanshahi
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada; Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Hong Ouyang
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Choudhary Ahmed
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ali Zahedi Amiri
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Subha Dahal
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Yu-Qian Mao
- Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada
| | | | - Ramy Malty
- Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada; Research and Innovation Centre, Department of Biochemistry, University of Regina, Regina, SK, Canada
| | - Wenming Duan
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Terek Been
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Maria Mangos
- Donnelly Center, University of Toronto, Ontario, Canada
| | | | - Mohan Babu
- Research and Innovation Centre, Department of Biochemistry, University of Regina, Regina, SK, Canada
| | - Liliana Attisano
- Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada; Donnelly Center, University of Toronto, Ontario, Canada
| | - Walid A Houry
- Dept. of Biochemistry, University of Toronto, Toronto, ON, Canada; Dept. of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Theo J Moraes
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Alan Cochrane
- Dept. of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
21
|
Mandal SK, Rehman MDMU, Katyal A, Rajvanshi K, Kannan M, Garg M, Murugesan S, Deepa P. In silico anti-viral assessment of phytoconstituents in a traditional (Siddha Medicine) polyherbal formulation - Targeting Mpro and pan-coronavirus post-fusion Spike protein. J Tradit Complement Med 2024; 14:55-69. [PMID: 38223813 PMCID: PMC10785248 DOI: 10.1016/j.jtcme.2023.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 01/16/2024] Open
Abstract
Background and aim Novel nature of the viral pathogen SARS-CoV-2 and the absence of standard drugs for treatment, have been a major challenge to combat this deadly infection. Natural products offer safe and effective remedy, for which traditional ethnic medicine can provide leads. An indigenous poly-herbal formulation, Kabasura Kudineer from Siddha system of medicine was evaluated here using a combination of computational approaches, to identify potential inhibitors against two anti-SARS-CoV-2 targets - post-fusion Spike protein (structural protein) and main protease (Mpro, non-structural protein). Experimental procedure We docked 32 phytochemicals from the poly-herbal formulation against viral post-fusion Spike glycoprotein and Mpro followed by molecular dynamics using Schrodinger software. Drug-likeness analysis was performed using machine learning (ML) approach and pkCSM. Results The binding affinity of the phytochemicals in Kabasura Kudineer revealed the following top-five bioactives: Quercetin > Luteolin > Chrysoeriol > 5-Hydroxy-7,8-Dimethoxyflavone > Scutellarein against Mpro target, and Gallic acid > Piperlonguminine > Chrysoeriol > Elemol > Piperine against post-fusion Spike protein target. Quercetin and Gallic acid exhibited binding stability in complexation with their respective viral-targets and favourable free energy change as revealed by the molecular dynamics simulations and MM-PBSA analysis. In silico predicted pharmacokinetic profiling of these ligands revealed appropriate drug-likeness properties. Conclusion These outcomes provide: (a) potential mechanism for the anti-viral efficacy of the indigenous Siddha formulation, targeting Mpro and post-fusion Spike protein (b) top bioactive lead-molecules that may be developed as natural product-based anti-viral pharmacotherapy and their pleiotropic protective effects may be leveraged to manage co-morbidities associated with COVID-19.
Collapse
Affiliation(s)
- Sumit Kumar Mandal
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| | - MD Muzaffar-Ur Rehman
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Ashish Katyal
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Kanishk Rajvanshi
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Manoj Kannan
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
- Plaksha University, SAS Nagar, Mohali, 140306, Punjab, India
| | - Mohit Garg
- Department of Chemical Engineering, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - P.R. Deepa
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| |
Collapse
|
22
|
Arman BY, Brun J, Hill ML, Zitzmann N, von Delft A. An Update on SARS-CoV-2 Clinical Trial Results-What We Can Learn for the Next Pandemic. Int J Mol Sci 2023; 25:354. [PMID: 38203525 PMCID: PMC10779148 DOI: 10.3390/ijms25010354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed over 7 million lives worldwide, providing a stark reminder of the importance of pandemic preparedness. Due to the lack of approved antiviral drugs effective against coronaviruses at the start of the pandemic, the world largely relied on repurposed efforts. Here, we summarise results from randomised controlled trials to date, as well as selected in vitro data of directly acting antivirals, host-targeting antivirals, and immunomodulatory drugs. Overall, repurposing efforts evaluating directly acting antivirals targeting other viral families were largely unsuccessful, whereas several immunomodulatory drugs led to clinical improvement in hospitalised patients with severe disease. In addition, accelerated drug discovery efforts during the pandemic progressed to multiple novel directly acting antivirals with clinical efficacy, including small molecule inhibitors and monoclonal antibodies. We argue that large-scale investment is required to prepare for future pandemics; both to develop an arsenal of broad-spectrum antivirals beyond coronaviruses and build worldwide clinical trial networks that can be rapidly utilised.
Collapse
Affiliation(s)
- Benediktus Yohan Arman
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Juliane Brun
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Michelle L. Hill
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
| | - Nicole Zitzmann
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Annette von Delft
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
- Centre for Medicine Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
23
|
Ahmad S, Xu J, Feng JA, Hutchinson A, Zeng H, Ghiabi P, Dong A, Centrella PA, Clark MA, Guié MA, Guilinger JP, Keefe AD, Zhang Y, Cerruti T, Cuozzo JW, von Rechenberg M, Bolotokova A, Li Y, Loppnau P, Seitova A, Li YY, Santhakumar V, Brown PJ, Ackloo S, Halabelian L. Discovery of a First-in-Class Small-Molecule Ligand for WDR91 Using DNA-Encoded Chemical Library Selection Followed by Machine Learning. J Med Chem 2023; 66:16051-16061. [PMID: 37996079 DOI: 10.1021/acs.jmedchem.3c01471] [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] [Indexed: 11/25/2023]
Abstract
WD40 repeat-containing protein 91 (WDR91) regulates early-to-late endosome conversion and plays vital roles in endosome fusion, recycling, and transport. WDR91 was recently identified as a potential host factor for viral infection. We employed DNA-encoded chemical library (DEL) selection against the WDR domain of WDR91, followed by machine learning to predict ligands from the synthetically accessible Enamine REAL database. Screening of predicted compounds identified a WDR91 selective compound 1, with a KD of 6 ± 2 μM by surface plasmon resonance. The co-crystal structure confirmed the binding of 1 to the WDR91 side pocket, in proximity to cysteine 487, which led to the discovery of covalent analogues 18 and 19. The covalent adduct formation for 18 and 19 was confirmed by intact mass liquid chromatography-mass spectrometry. The discovery of 1, 18, and 19, accompanying structure-activity relationship, and the co-crystal structures provide valuable insights for designing potent and selective chemical tools against WDR91 to evaluate its therapeutic potential.
Collapse
Affiliation(s)
- Shabbir Ahmad
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Jin Xu
- Google Research, Mountain View, California 94043, United States
| | - Jianwen A Feng
- Google Research, Mountain View, California 94043, United States
| | - Ashley Hutchinson
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Hong Zeng
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Pegah Ghiabi
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Aiping Dong
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Paolo A Centrella
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02435, United States
| | - Matthew A Clark
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02435, United States
| | - Marie-Aude Guié
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02435, United States
| | - John P Guilinger
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02435, United States
| | - Anthony D Keefe
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02435, United States
| | - Ying Zhang
- X-Chem Inc., 100 Beaver Street, Waltham, Massachusetts 02435, United States
| | - Thomas Cerruti
- Relay Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02139, United States
| | - John W Cuozzo
- Relay Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02139, United States
| | - Moritz von Rechenberg
- Relay Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02139, United States
| | - Albina Bolotokova
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Yanjun Li
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Peter Loppnau
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Alma Seitova
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Yen-Yen Li
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | | | - Peter J Brown
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Suzanne Ackloo
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
| | - Levon Halabelian
- Structural Genomics Consortium, University of Toronto, Ontario M5G 1L7, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto M5S 1A8, Ontario, Canada
| |
Collapse
|
24
|
Stewart H, Palmulli R, Johansen KH, McGovern N, Shehata OM, Carnell GW, Jackson HK, Lee JS, Brown JC, Burgoyne T, Heeney JL, Okkenhaug K, Firth AE, Peden AA, Edgar JR. Tetherin antagonism by SARS-CoV-2 ORF3a and spike protein enhances virus release. EMBO Rep 2023; 24:e57224. [PMID: 37818801 PMCID: PMC10702813 DOI: 10.15252/embr.202357224] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/23/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023] Open
Abstract
The antiviral restriction factor, tetherin, blocks the release of several different families of enveloped viruses, including the Coronaviridae. Tetherin is an interferon-induced protein that forms parallel homodimers between the host cell and viral particles, linking viruses to the surface of infected cells and inhibiting their release. We demonstrate that SARS-CoV-2 infection causes tetherin downregulation and that tetherin depletion from cells enhances SARS-CoV-2 viral titres. We investigate the potential viral proteins involved in abrogating tetherin function and find that SARS-CoV-2 ORF3a reduces tetherin localisation within biosynthetic organelles where Coronaviruses bud, and increases tetherin localisation to late endocytic organelles via reduced retrograde recycling. We also find that expression of Spike protein causes a reduction in cellular tetherin levels. Our results confirm that tetherin acts as a host restriction factor for SARS-CoV-2 and highlight the multiple distinct mechanisms by which SARS-CoV-2 subverts tetherin function.
Collapse
Affiliation(s)
- Hazel Stewart
- Department of PathologyUniversity of CambridgeCambridgeUK
| | | | - Kristoffer H Johansen
- Department of PathologyUniversity of CambridgeCambridgeUK
- Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMDUSA
| | - Naomi McGovern
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - Ola M Shehata
- Department of Biomedical ScienceUniversity of Sheffield, Firth CourtSheffieldUK
| | - George W Carnell
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | | | - Jin S Lee
- Department of PathologyUniversity of CambridgeCambridgeUK
| | | | - Thomas Burgoyne
- Royal Brompton HospitalGuy's and St Thomas' NHS Foundation TrustLondonUK
- UCL Institute of OphthalmologyUniversity College LondonLondonUK
| | | | | | - Andrew E Firth
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - Andrew A Peden
- Department of Biomedical ScienceUniversity of Sheffield, Firth CourtSheffieldUK
| | - James R Edgar
- Department of PathologyUniversity of CambridgeCambridgeUK
| |
Collapse
|
25
|
Dasgupta A, Gangai S, Narayan R, Kapoor S. Mapping the Lipid Signatures in COVID-19 Infection: Diagnostic and Therapeutic Solutions. J Med Chem 2023; 66:14411-14433. [PMID: 37899546 DOI: 10.1021/acs.jmedchem.3c01238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
The COVID-19 pandemic ignited research centered around the identification of robust biomarkers and therapeutic targets. SARS-CoV-2, the virus responsible, hijacks the metabolic machinery of the host cells. It relies on lipids and lipoproteins of host cells for entry, trafficking, immune evasion, viral replication, and exocytosis. The infection causes host cell lipid metabolic remodelling. Targeting lipid-based processes is thus a promising strategy for countering COVID-19. Here, we review the role of lipids in the different steps of the SARS-CoV-2 pathogenesis and identify lipid-centric targetable avenues. We discuss lipidome changes in infected patients and their relevance as potential clinical diagnostic or prognostic biomarkers. We summarize the emerging direct and indirect therapeutic approaches for targeting COVID-19 using lipid-inspired approaches. Given that viral protein-targeted therapies may become less effective due to mutations in emerging SARS-CoV-2 variants, lipid-inspired interventions may provide additional and perhaps better means of combating this and future pandemics.
Collapse
Affiliation(s)
- Aishi Dasgupta
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
- IIT-Bombay Monash Academy, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shon Gangai
- School of Chemical and Materials Sciences (SCMS), Institute of Technology Goa, Farmagudi, Ponda, Goa 403401, India
| | - Rishikesh Narayan
- School of Chemical and Materials Sciences (SCMS), Institute of Technology Goa, Farmagudi, Ponda, Goa 403401, India
- School of Interdisciplinary Life Sciences (SILS), Institute of Technology Goa, Farmagudi, Ponda, Goa 403401, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
- IIT-Bombay Monash Academy, Indian Institute of Technology Bombay, Mumbai 400076, India
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| |
Collapse
|
26
|
Sayaf AM, Ahmad H, Aslam MA, Ghani SA, Bano S, Yousafi Q, Suleman M, Khan A, Yeoh KK, Wei DQ. Pharmacotherapeutic Potential of Natural Products to Target the SARS-CoV-2 PLpro Using Molecular Screening and Simulation Approaches. Appl Biochem Biotechnol 2023; 195:6959-6978. [PMID: 36961512 PMCID: PMC10037394 DOI: 10.1007/s12010-023-04466-1] [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] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
Because of the essential role of PLpro in the regulation of replication and dysregulation of the host immune sensing, it is considered a therapeutic target for novel drug development. To reduce the risk of immune evasion and vaccine effectiveness, small molecular therapeutics are the best complementary approach. Hence, we used a structure-based drug-designing approach to identify potential small molecular inhibitors for PLpro of SARS-CoV-2. Initial scoring and re-scoring of the best hits revealed that three compounds NPC320891 (2,2-Dihydroxyindene-1,3-Dione), NPC474594 (Isonarciclasine), and NPC474595 (7-Deoxyisonarciclasine) exhibit higher docking scores than the control GRL0617. Investigation of the binding modes revealed that alongside the essential contacts, i.e., Asp164, Glu167, Tyr264, and Gln269, these molecules also target Lys157 and Tyr268 residues in the active site. Moreover, molecular simulation demonstrated that the reported top hits also possess stable dynamics and structural packing. Furthermore, the residues' flexibility revealed that all the complexes demonstrated higher flexibility in the regions 120-140, 160-180, and 205-215. The 120-140 and 160-180 lie in the finger region of PLpro, which may open/close during the simulation to cover the active site and push the ligand inside. In addition, the total binding free energy was reported to be - 32.65 ± 0.17 kcal/mol for the GRL0617-PLpro, for the NPC320891-PLpro complex, the TBE was - 35.58 ± 0.14 kcal/mol, for the NPC474594-PLpro, the TBE was - 43.72 ± 0.22 kcal/mol, while for NPC474595-PLpro complex, the TBE was calculated to be - 41.61 ± 0.20 kcal/mol, respectively. Clustering of the protein's motion and FEL further revealed that in NPC474594 and NPC474595 complexes, the drug was seen to have moved inside the binding cavity along with the loop in the palm region harboring the catalytic triad, thus justifying the higher binding of these two molecules particularly. In conclusion, the overall results reflect favorable binding of the identified hits strongly than the control drug, thus demanding in vitro and in vivo validation for clinical purposes.
Collapse
Affiliation(s)
- Abrar Mohammad Sayaf
- School of Chemical Sciences, Universiti Sains Malaysia, 11800 USM George Town, Penang Malaysia
| | - Hassaan Ahmad
- Rawalpindi Medical University, Chamanzar Colony, Rawalpindi, Punjab 46000, Pakistan
| | - Muhammad Ammar Aslam
- Rawalpindi Medical University, Chamanzar Colony, Rawalpindi, Punjab 46000, Pakistan
| | | | - Saira Bano
- Department of Botany, University of Okara, Okara, Punjab Pakistan
| | - Qudsia Yousafi
- Department of Biosciences, COMSATS University Islamabad-Sahiwal Campus, Sahiwal, Punjab Pakistan
| | - Muhammad Suleman
- Centre for Biotechnology and Microbiology, University of Swat, Swat, Khyber Pakhtunkhwa Pakistan
| | - Abbas Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240 People’s Republic of China
- Zhongjing Research and Industrialization, Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang, Henan 473006 People’s Republic of China
| | - Kar Kheng Yeoh
- School of Chemical Sciences, Universiti Sains Malaysia, 11800 USM George Town, Penang Malaysia
| | - Dong-Qing Wei
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240 People’s Republic of China
- Zhongjing Research and Industrialization, Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang, Henan 473006 People’s Republic of China
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center On Antibacterial Resistances, Joint Laboratory of International Laboratory of Metabolic and Developmental Sciences, Ministry of Education and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200030 People’s Republic of China
- Peng Cheng Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nanshan District, Shenzhen, Guangdong 518055 People’s Republic of China
| |
Collapse
|
27
|
Malireddi RKS, Bynigeri RR, Mall R, Connelly JP, Pruett-Miller SM, Kanneganti TD. Inflammatory cell death, PANoptosis, screen identifies host factors in coronavirus innate immune response as therapeutic targets. Commun Biol 2023; 6:1071. [PMID: 37864059 PMCID: PMC10589293 DOI: 10.1038/s42003-023-05414-9] [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: 05/22/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023] Open
Abstract
The COVID-19 pandemic, caused by the β-coronavirus (β-CoV) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), continues to cause significant global morbidity and mortality. While vaccines have reduced the overall number of severe infections, there remains an incomplete understanding of viral entry and innate immune activation, which can drive pathology. Innate immune responses characterized by positive feedback between cell death and cytokine release can amplify the inflammatory cytokine storm during β-CoV-mediated infection to drive pathology. Therefore, there remains an unmet need to understand innate immune processes in response to β-CoV infections to identify therapeutic strategies. To address this gap, here we used an MHV model and developed a whole genome CRISPR-Cas9 screening approach to elucidate host molecules required for β-CoV infection and inflammatory cell death, PANoptosis, in macrophages, a sentinel innate immune cell. Our screen was validated through the identification of the known MHV receptor Ceacam1 as the top hit, and its deletion significantly reduced viral replication due to loss of viral entry, resulting in a downstream reduction in MHV-induced cell death. Moreover, this screen identified several other host factors required for MHV infection-induced macrophage cell death. Overall, these findings demonstrate the feasibility and power of using genome-wide PANoptosis screens in macrophage cell lines to accelerate the discovery of key host factors in innate immune processes and suggest new targets for therapeutic development to prevent β-CoV-induced pathology.
Collapse
Affiliation(s)
- R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ratnakar R Bynigeri
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Raghvendra Mall
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Biotechnology Research Center, Technology Innovation Institute, Abu Dhabi, P.O. Box 9639, United Arab Emirates
| | - Jon P Connelly
- Center for Advanced Genome Engineering (CAGE), St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering (CAGE), St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | |
Collapse
|
28
|
Anesi GL, Degnan K, Dutcher L, Saw S, Maguire C, Binkley A, Patel S, Athans V, Barton TD, Binkley S, Candeloro CL, Herman DJ, Kasbekar N, Kennedy L, Millstein JH, Meyer NJ, Talati NJ, Patel H, Pegues DA, Sayre PJ, Tebas P, Terico AT, Murphy KM, O’Donnell JA, White M, Hamilton KW. The Penn Medicine COVID-19 Therapeutics Committee-Reflections on a Model for Rapid Evidence Review and Dynamic Practice Recommendations During a Public Health Emergency. Open Forum Infect Dis 2023; 10:ofad428. [PMID: 37663091 PMCID: PMC10468749 DOI: 10.1093/ofid/ofad428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
The Penn Medicine COVID-19 Therapeutics Committee-an interspecialty, clinician-pharmacist, and specialist-front line primary care collaboration-has served as a forum for rapid evidence review and the production of dynamic practice recommendations during the 3-year coronavirus disease 2019 public health emergency. We describe the process by which the committee went about its work and how it navigated specific challenging scenarios. Our target audiences are clinicians, hospital leaders, public health officials, and researchers invested in preparedness for inevitable future threats. Our objectives are to discuss the logistics and challenges of forming an effective committee, undertaking a rapid evidence review process, aligning evidence-based guidelines with operational realities, and iteratively revising recommendations in response to changing pandemic data. We specifically discuss the arc of evidence for corticosteroids; the noble beginnings and dangerous misinformation end of hydroxychloroquine and ivermectin; monoclonal antibodies and emerging viral variants; and patient screening and safety processes for tocilizumab, baricitinib, and nirmatrelvir-ritonavir.
Collapse
Affiliation(s)
- George L Anesi
- Division of Pulmonary, Allergy, and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kathleen Degnan
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lauren Dutcher
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Stephen Saw
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Christina Maguire
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Amanda Binkley
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Sonal Patel
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Vasilios Athans
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Todd D Barton
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shawn Binkley
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Christina L Candeloro
- Department of Pharmacy, Hospital of the University of Pennsylvania, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - David J Herman
- Division of Infectious Diseases, Penn Medicine Princeton Medical Center, University of Pennsylvania Health System, Princeton, New Jersey, USA
| | - Nishaminy Kasbekar
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Leigh Kennedy
- Division of Infectious Diseases, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Jeffrey H Millstein
- Regional Physician Practices of Penn Medicine, Woodbury Heights, New Jersey, USA
| | - Nuala J Meyer
- Division of Pulmonary, Allergy, and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Naasha J Talati
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hinal Patel
- Department of Pharmacy, Penn Medicine Princeton Medical Center, University of Pennsylvania Health System, Princeton, New Jersey, USA
| | - David A Pegues
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Patrick J Sayre
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Pablo Tebas
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Adrienne T Terico
- Department of Pharmacy, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA
| | - Kathleen M Murphy
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Judith A O’Donnell
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Melissa White
- Department of Pharmacy, Penn Medicine Lancaster General Health, University of Pennsylvania Health System, Lancaster, Pennsylvania, USA
| | - Keith W Hamilton
- Division of Infectious Diseases, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
Zhou C, Liu Y, Wei Q, Chen Y, Yang S, Cheng A, Zhang G. HSPA5 Promotes Attachment and Internalization of Porcine Epidemic Diarrhea Virus through Interaction with the Spike Protein and the Endo-/Lysosomal Pathway. J Virol 2023; 97:e0054923. [PMID: 37222617 PMCID: PMC10308931 DOI: 10.1128/jvi.00549-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/25/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) has caused huge economic losses to the global pig industry. The swine enteric coronavirus spike (S) protein recognizes various cell surface molecules to regulate viral infection. In this study, we identified 211 host membrane proteins related to the S1 protein by pulldown combined with liquid-chromatography tandem mass spectrometry (LC-MS/MS) analysis. Among these, heat shock protein family A member 5 (HSPA5) was identified through screening as having a specific interaction with the PEDV S protein, and positive regulation of PEDV infection was validated by knockdown and overexpression tests. Further studies verified the role of HSPA5 in viral attachment and internalization. In addition, we found that HSPA5 interacts with S proteins through its nucleotide-binding structural domain (NBD) and that polyclonal antibodies can block viral infection. In detail, HSPA5 was found to be involved in viral trafficking via the endo-/lysosomal pathway. Inhibition of HSPA5 activity during internalization would reduce the subcellular colocalization of PEDV with lysosomes in the endo-/lysosomal pathway. Together, these findings show that HSPA5 is a novel PEDV potential target for the creation of therapeutic drugs. IMPORTANCE PEDV infection causes severe piglet mortality and threatens the global pig industry. However, the complex invasion mechanism of PEDV makes its prevention and control difficult. Here, we determined that HSPA5 is a novel target for PEDV which interacts with its S protein and is involved in viral attachment and internalization, influencing its transport via the endo-/lysosomal pathway. Our work extends knowledge about the relationship between the PEDV S and host proteins and provides a new therapeutic target against PEDV infection.
Collapse
Affiliation(s)
- Chuanjie Zhou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Yunchao Liu
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Qiang Wei
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Yumei Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Suzhen Yang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Anchun Cheng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
30
|
Chen XN, Liang YF, Weng ZJ, Quan WP, Hu C, Peng YZ, Sun YS, Gao Q, Huang Z, Zhang GH, Gong L. Porcine Enteric Alphacoronavirus Entry through Multiple Pathways (Caveolae, Clathrin, and Macropinocytosis) Requires Rab GTPases for Endosomal Transport. J Virol 2023; 97:e0021023. [PMID: 36975780 PMCID: PMC10134835 DOI: 10.1128/jvi.00210-23] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Porcine enteric alphacoronavirus (PEAV) is a new bat HKU2-like porcine coronavirus, and its endemic outbreak has caused severe economic losses to the pig industry. Its broad cellular tropism suggests a potential risk of cross-species transmission. A limited understanding of PEAV entry mechanisms may hinder a rapid response to potential outbreaks. This study analyzed PEAV entry events using chemical inhibitors, RNA interference, and dominant-negative mutants. PEAV entry into Vero cells depended on three endocytic pathways: caveolae, clathrin, and macropinocytosis. Endocytosis requires dynamin, cholesterol, and a low pH. Rab5, Rab7, and Rab9 GTPases (but not Rab11) regulate PEAV endocytosis. PEAV particles colocalize with EEA1, Rab5, Rab7, Rab9, and Lamp-1, suggesting that PEAV translocates into early endosomes after internalization, and Rab5, Rab7, and Rab9 regulate trafficking to lysosomes before viral genome release. PEAV enters porcine intestinal cells (IPI-2I) through the same endocytic pathway, suggesting that PEAV may enter various cells through multiple endocytic pathways. This study provides new insights into the PEAV life cycle. IMPORTANCE Emerging and reemerging coronaviruses cause severe human and animal epidemics worldwide. PEAV is the first bat-like coronavirus to cause infection in domestic animals. However, the PEAV entry mechanism into host cells remains unknown. This study demonstrates that PEAV enters into Vero or IPI-2I cells through caveola/clathrin-mediated endocytosis and macropinocytosis, which does not require a specific receptor. Subsequently, Rab5, Rab7, and Rab9 regulate PEAV trafficking from early endosomes to lysosomes, which is pH dependent. The results advance our understanding of the disease and help to develop potential new drug targets against PEAV.
Collapse
Affiliation(s)
- Xiong-nan Chen
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Yi-fan Liang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Zhi-jun Weng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Wei-peng Quan
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, People’s Republic of China
| | - Chen Hu
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Yun-zhao Peng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, People’s Republic of China
| | - Ying-shuo Sun
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Qi Gao
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Zhao Huang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Gui-hong Zhang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, People’s Republic of China
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Lang Gong
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, People’s Republic of China
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, People’s Republic of China
| |
Collapse
|
31
|
Sun Q, Li X, Kuang E. Subversion of autophagy machinery and organelle-specific autophagy by SARS-CoV-2 and coronaviruses. Autophagy 2023; 19:1055-1069. [PMID: 36005882 PMCID: PMC10012907 DOI: 10.1080/15548627.2022.2116677] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 12/09/2022] Open
Abstract
As a new emerging severe coronavirus, the knowledge on the SARS-CoV-2 and COVID-19 remains very limited, whereas many concepts can be learned from the homologous coronaviruses. Macroautophagy/autophagy is finely regulated by SARS-CoV-2 infection and plays important roles in SARS-CoV-2 infection and pathogenesis. This review will explore the subversion and mechanism of the autophagy-related machinery, vacuoles and organelle-specific autophagy during infection of SARS-CoV-2 and coronaviruses to provide meaningful insights into the autophagy-related therapeutic strategies for infectious diseases of SARS-CoV-2 and coronaviruses.
Collapse
Affiliation(s)
- Qinqin Sun
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaojuan Li
- College of Clinic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Ersheng Kuang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Ministry of Education, Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Guangzhou, Guangdong, China
| |
Collapse
|
32
|
Liu Y, Zhang Q, Zou M, Cui J, Shi X, Li L, Wu F, Xu X. Cell entry of Bovine herpesvirus-1 through clathrin- and caveolin-mediated endocytosis requires activation of PI3K-Akt-NF-κB and Ras-p38 MAPK pathways as well as the interaction of BoHV-1 gD with cellular receptor nectin-1. Vet Microbiol 2023; 279:109672. [PMID: 36774841 DOI: 10.1016/j.vetmic.2023.109672] [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: 12/06/2022] [Revised: 01/17/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Bovine herpesvirus-1 (BoHV-1) can infect all breeds of cattle and cause severe respiratory organs and genital tract diseases. However, the mechanism of BoHV-1 entering the cells remains unclear. In this study, we explored the mechanism of BoHV-1 entering MDBK cells. We found that the entry of BoHV-1 was blocked by NH4Cl and bafilomycin A1, indicating that BoHV-1 entry is dependent on the acidic environment of endosome. Specific inhibitor dynasore and small interfering RNA (siRNA) knockdown of dynamin-2 inhibited BoHV-1 entry, showing that dynamin is required in BoHV-1 entry. The results of specific inhibitor, siRNA knockdown and co-localization indicating clathrin- and caveolin- mediated endocytosis play a role in BoHV-1 entry. BoHV-1 infection was not affected by EIPA which is a specific inhibitor of macropinocytosis. In addition, we found that BoHV-1 triggered PI3K-Akt-NF-κB and Ras-p38 MAPK signaling pathways to induce clathrin-mediated and caveolin-mediated endocytosis at the early stage of BoHV-1 infection. BoHV-1 binding was sufficient to activate the endocytic signaling pathways and promote viral entry. These two signaling pathways were activated by transfection of viral gD protein, and were inhibited by deletion of viral gD protein and the siRNA knockdown of cellular receptor nectin-1. The results of co-localization indicating the entered BoHV-1 is traced to late endosomes via early endosomes. Our results suggested the interaction of viral gD protein and cellular receptor nectin-1 triggered the PI3K-Akt-NF-κB and Ras-p38 MAPK signaling pathways and induced clathrin-mediated and caveolin-mediated endocytosis to promote BoHV-1 entry into MDBK cells at the early stage of BoHV-1 infection.
Collapse
Affiliation(s)
- Yang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Animal Biosafety Risk Warning and Control of Ministry of Agriculture and Rural Affairs (South), China Animal Health And Epidemiology Center, Qingdao, Shandong 266032, China
| | - Qi Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Min Zou
- State Key Laboratory of Animal Genetical Engineered Vaccine of Ministry of Science and Technology, Qingdao YeBio Biological Engineering Company Limited, Qingdao, Shandong 266110, China
| | - Jin Cui
- Key Laboratory of Animal Biosafety Risk Warning and Control of Ministry of Agriculture and Rural Affairs (South), China Animal Health And Epidemiology Center, Qingdao, Shandong 266032, China
| | - Xiaojie Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Linjie Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Faxing Wu
- Key Laboratory of Animal Biosafety Risk Warning and Control of Ministry of Agriculture and Rural Affairs (South), China Animal Health And Epidemiology Center, Qingdao, Shandong 266032, China.
| | - Xingang Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
33
|
Contreras PS, Tapia PJ, Jeong E, Ghosh S, Altan-Bonnet N, Puertollano R. Beta-coronaviruses exploit cellular stress responses by modulating TFEB and TFE3 activity. iScience 2023; 26:106169. [PMID: 36785787 PMCID: PMC9908431 DOI: 10.1016/j.isci.2023.106169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/09/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Beta-coronaviruses have emerged as a severe threat to global health. Undercovering the interplay between host and beta-coronaviruses is essential for understanding disease pathogenesis and developing efficient treatments. Here we report that the transcription factors TFEB and TFE3 translocate from the cytosol to the nucleus in response to beta-coronavirus infection by a mechanism that requires activation of calcineurin phosphatase. In the nucleus, TFEB and TFE3 bind to the promoter of multiple lysosomal and immune genes. Accordingly, MHV-induced upregulation of immune regulators is significantly decreased in TFEB/TFE3-depleted cells. Conversely, over-expression of either TFEB or TFE3 is sufficient to increase expression of several cytokines and chemokines. The reduced immune response observed in the absence of TFEB and TFE3 results in increased cellular survival of infected cells but also in reduced lysosomal exocytosis and decreased viral infectivity. These results suggest a central role of TFEB and TFE3 in cellular response to beta-coronavirus infection.
Collapse
Affiliation(s)
- Pablo S. Contreras
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pablo J. Tapia
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eutteum Jeong
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sourish Ghosh
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nihal Altan-Bonnet
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
34
|
Roy SS, Sharma S, Rizvi ZA, Sinha D, Gupta D, Rophina M, Sehgal P, Sadhu S, Tripathy MR, Samal S, Maiti S, Scaria V, Sivasubbu S, Awasthi A, Harshan KH, Jain S, Chowdhury S. G4-binding drugs, chlorpromazine and prochlorperazine, repurposed against COVID-19 infection in hamsters. Front Mol Biosci 2023; 10:1133123. [PMID: 37006620 PMCID: PMC10061221 DOI: 10.3389/fmolb.2023.1133123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has caused millions of infections and deaths worldwide. Limited treatment options and the threat from emerging variants underline the need for novel and widely accessible therapeutics. G-quadruplexes (G4s) are nucleic acid secondary structures known to affect many cellular processes including viral replication and transcription. We identified heretofore not reported G4s with remarkably low mutation frequency across >5 million SARS-CoV-2 genomes. The G4 structure was targeted using FDA-approved drugs that can bind G4s - Chlorpromazine (CPZ) and Prochlorperazine (PCZ). We found significant inhibition in lung pathology and lung viral load of SARS-CoV-2 challenged hamsters when treated with CPZ or PCZ that was comparable to the widely used antiviral drug Remdesivir. In support, in vitro G4 binding, inhibition of reverse transcription from RNA isolated from COVID-infected humans, and attenuated viral replication and infectivity in Vero cell cultures were clear in case of both CPZ and PCZ. Apart from the wide accessibility of CPZ/PCZ, targeting relatively invariant nucleic acid structures poses an attractive strategy against viruses like SARS-CoV-2, which spread fast and accumulate mutations quickly.
Collapse
Affiliation(s)
- Shuvra Shekhar Roy
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shalu Sharma
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Zaigham Abbas Rizvi
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Dipanjali Sinha
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Divya Gupta
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Mercy Rophina
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Paras Sehgal
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srikanth Sadhu
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Manas Ranjan Tripathy
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Sweety Samal
- Translational Health Science and Technology Institute, Faridabad, 411008, India
| | - Souvik Maiti
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- CSIR-National Chemical Laboratory, Pune, 121001, India
| | - Vinod Scaria
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Amit Awasthi
- Immuno-biology Laboratory, Infection and Immunology Centre, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Krishnan H. Harshan
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Sanjeev Jain
- Molecular Genetics Laboratory, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Shantanu Chowdhury
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, 110025, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
- *Correspondence: Shantanu Chowdhury,
| |
Collapse
|
35
|
Gelinas AD, Tan TK, Liu S, Jaramillo JG, Chadwick J, Harding AC, Zhang C, Ream BE, Chase CN, Otis MR, Lee T, Schneider DJ, James WS, Janjic N. Broadly neutralizing aptamers to SARS-CoV-2: A diverse panel of modified DNA antiviral agents. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:370-382. [PMID: 36714461 PMCID: PMC9859636 DOI: 10.1016/j.omtn.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Since its discovery, COVID-19 has rapidly spread across the globe and has had a massive toll on human health, with infection mortality rates as high as 10%, and a crippling impact on the world economy. Despite numerous advances, there remains an urgent need for accurate and rapid point-of-care diagnostic tests and better therapeutic treatment options. To contribute chemically distinct, non-protein-based affinity reagents, we report here the identification of modified DNA-based aptamers that selectively bind to the S1, S2, or receptor-binding domain of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. Several aptamers inhibit the binding of the spike protein to its cell-surface receptor angiotensin-converting enzyme 2 (ACE2) and neutralize authentic SARS-CoV-2 virus in vitro, including all variants of concern. With a high degree of nuclease resistance imparted by the base modifications, these reagents represent a new class of molecules with potential for further development as diagnostics or therapeutics.
Collapse
Affiliation(s)
- Amy D. Gelinas
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO 80301, USA
| | - Tiong Kit Tan
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Sai Liu
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Javier G. Jaramillo
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - James Chadwick
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Adam C. Harding
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Chi Zhang
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO 80301, USA
| | - Brian E. Ream
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO 80301, USA
| | | | - Matthew R. Otis
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO 80301, USA
| | - Thomas Lee
- Department of Biochemistry, University of Colorado, Boulder, JSCBB, C1B90, 3415 Colorado Avenue, Boulder, CO 80303, USA
| | | | - William S. James
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
- Corresponding author William S. James, James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| | - Nebojsa Janjic
- SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO 80301, USA
- Corresponding author Nebojsa Janjic, SomaLogic, Inc., 2945 Wilderness Place, Boulder, CO 80301, USA.
| |
Collapse
|
36
|
Doijen J, Temmerman K, Van den Eynde C, Diels A, Van den Broeck N, Van Gool M, Heo I, Jaensch S, Zwaagstra M, Diosa Toro M, Chiu W, De Jonghe S, Leyssen P, Bojkova D, Ciesek S, Cinatl J, Verschueren L, Buyck C, Van Kuppeveld F, Neyts J, Van Loock M, Van Damme E. Identification of Z-Tyr-Ala-CHN 2, a Cathepsin L Inhibitor with Broad-Spectrum Cell-Specific Activity against Coronaviruses, including SARS-CoV-2. Microorganisms 2023; 11:717. [PMID: 36985290 PMCID: PMC10055926 DOI: 10.3390/microorganisms11030717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/17/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The ongoing COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is partly under control by vaccination. However, highly potent and safe antiviral drugs for SARS-CoV-2 are still needed to avoid development of severe COVID-19. We report the discovery of a small molecule, Z-Tyr-Ala-CHN2, which was identified in a cell-based antiviral screen. The molecule exerts sub-micromolar antiviral activity against SARS-CoV-2, SARS-CoV-1, and human coronavirus 229E. Time-of-addition studies reveal that Z-Tyr-Ala-CHN2 acts at the early phase of the infection cycle, which is in line with the observation that the molecule inhibits cathepsin L. This results in antiviral activity against SARS-CoV-2 in VeroE6, A549-hACE2, and HeLa-hACE2 cells, but not in Caco-2 cells or primary human nasal epithelial cells since the latter two cell types also permit entry via transmembrane protease serine subtype 2 (TMPRSS2). Given their cell-specific activity, cathepsin L inhibitors still need to prove their value in the clinic; nevertheless, the activity profile of Z-Tyr-Ala-CHN2 makes it an interesting tool compound for studying the biology of coronavirus entry and replication.
Collapse
Affiliation(s)
- Jordi Doijen
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Koen Temmerman
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Annick Diels
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | | | - Inha Heo
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Steffen Jaensch
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Marleen Zwaagstra
- Faculty of Veterinary Medicine, Yalelaan 1, Virology Division, Department of Biomolecular Health Sciences, Infectious Diseases and Immunology, Utrecht University, 3584 Utrecht, The Netherlands
| | - Mayra Diosa Toro
- Faculty of Veterinary Medicine, Yalelaan 1, Virology Division, Department of Biomolecular Health Sciences, Infectious Diseases and Immunology, Utrecht University, 3584 Utrecht, The Netherlands
| | - Winston Chiu
- Laboratory of Virology and Chemotherapy, Herestraat 49, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Steven De Jonghe
- Laboratory of Virology and Chemotherapy, Herestraat 49, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Pieter Leyssen
- Laboratory of Virology and Chemotherapy, Herestraat 49, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Denisa Bojkova
- Institute for Medical Virology, University Hospital, Paul-Ehrlich-Str. 40, Frankfurt University, 60596 Frankfurt am Main, Germany
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital, Paul-Ehrlich-Str. 40, Frankfurt University, 60596 Frankfurt am Main, Germany
| | - Jindrich Cinatl
- Institute for Medical Virology, University Hospital, Paul-Ehrlich-Str. 40, Frankfurt University, 60596 Frankfurt am Main, Germany
| | - Lore Verschueren
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Christophe Buyck
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Frank Van Kuppeveld
- Faculty of Veterinary Medicine, Yalelaan 1, Virology Division, Department of Biomolecular Health Sciences, Infectious Diseases and Immunology, Utrecht University, 3584 Utrecht, The Netherlands
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Herestraat 49, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Marnix Van Loock
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Ellen Van Damme
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| |
Collapse
|
37
|
Rlip Protein: A Potential Target for COVID-19. J Community Hosp Intern Med Perspect 2023; 12:89-94. [PMID: 36816155 PMCID: PMC9924642 DOI: 10.55729/2000-9666.1090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/27/2022] [Accepted: 05/31/2022] [Indexed: 11/12/2022] Open
Abstract
On January 30, 2020, the COVID-19 epidemic was declared an international public health emergency by the World Health Organization. Given the growing impact of the pandemic, there is great interest in finding potential targets for treating infected or hospitalized COVID-19 patients. Therapeutic studies have been conducted on pre-existing drugs, which vary by country, including anti-malarial agents, antiviral agents, and convalescent plasma. However, many of these agents are ineffective at reducing mortality or only shorten the severity or duration of COVID-19 illness in hospitalized patients. As such, other alternatives for treating COVID-19 are being investigated. One such target of interest has been clathrin-dependent endocytosis (CDE). Clathrin-dependent endocytosis is the most commonly observed mechanism of viral entry into cells. However, there have been no published studies to date on CDE inhibition strategies against COVID-19. One such target is Rlip or RLIP76 (human gene RALBP1, 18p11.22). Among its many functions, Rlip is a stress-protective, Ral-regulated ATPase of the mercapturic acid pathway that transports glutathione-electrophile conjugates of electrophilic toxins, which are precursors of mercapturic acid that precedes de-glutamylation by gamma-glutamyl transferase. Rlip is also regulated by several G-proteins that coordinate movement of cells, organelles, membranes, cytoskeleton, macromolecules, and other small molecules. Previous studies have link Rlip in the pathogenesis of several viral illness. In this paper, we want to propose that RLIP76 (Rlip or RALBP1) may be a novel target for treating SARS-CoV-2 viral infections.
Collapse
|
38
|
Wang Y, Huang H, Li D, Zhao C, Li S, Qin P, Li Y, Yang X, Du W, Li W, Li Y. Identification of niclosamide as a novel antiviral agent against porcine epidemic diarrhea virus infection by targeting viral internalization. Virol Sin 2023; 38:296-308. [PMID: 36702255 PMCID: PMC10176444 DOI: 10.1016/j.virs.2023.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), an enteropathogenic coronavirus, has catastrophic impacts on the global pig industry. However, there remain no effective drugs against PEDV infection. In this study, we utilized a recombinant PEDV expressing renilla luciferase (PEDV-Rluc) to screen potential anti-PEDV agents from an FDA-approved drug library in Vero cells. Four compounds were identified that significantly decreased luciferase activity of PEDV-Rluc. Among them, niclosamide was further characterized because it exhibited the most potent antiviral activity with the highest selectivity index. It can efficiently inhibit viral RNA synthesis, protein expression and viral progeny production of classical and variant PEDV strains in a dose-dependent manner. Time of addition assay showed that niclosamide exhibited potent anti-PEDV activity when added simultaneously with or after virus infection. Furthermore, niclosamide significantly inhibited the entry stage of PEDV infection by affecting viral internalization rather than viral attachment to cells. In addition, a combination with other small molecule inhibitors of endosomal acidification enhanced the anti-PEDV effect of niclosamide in vitro. Taken together, these findings suggested that niclosamide is a novel antiviral agent that might provide a basis for the development of novel drug therapies against PEDV and other related pathogenic coronavirus infections.
Collapse
Affiliation(s)
- Yue Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Huimin Huang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Dongliang Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Chenxu Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Shuai Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Panpan Qin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Yaqin Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xia Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Wenjuan Du
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, the Netherlands
| | - Wentao Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China.
| | - Yongtao Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China; Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, the Netherlands.
| |
Collapse
|
39
|
Xing Y, Zhang Q, Jiu Y. Coronavirus and the Cytoskeleton of Virus-Infected Cells. Subcell Biochem 2023; 106:333-364. [PMID: 38159233 DOI: 10.1007/978-3-031-40086-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
The cytoskeleton, which includes actin filaments, microtubules, and intermediate filaments, is one of the most important networks in the cell and undertakes many fundamental life activities. Among them, actin filaments are mainly responsible for maintaining cell shape and mediating cell movement, microtubules are in charge of coordinating all cargo transport within the cell, and intermediate filaments are mainly thought to guard against external mechanical pressure. In addition to this, cytoskeleton networks are also found to play an essential role in multiple viral infections. Due to the COVID-19 epidemic, including SARS-CoV-2, SARS-CoV and MERS-CoV, so many variants have caused wide public concern, that any virus infection can potentially bring great harm to human beings and society. Therefore, it is of great importance to study coronavirus infection and develop antiviral drugs and vaccines. In this chapter, we summarize in detail how the cytoskeleton responds and participates in coronavirus infection by analyzing the possibility of the cytoskeleton and its related proteins as antiviral targets, thereby providing ideas for finding more effective treatments.
Collapse
Affiliation(s)
- Yifan Xing
- Shanghai Institute of Immunity and Infection (Formerly Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian Zhang
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yaming Jiu
- Shanghai Institute of Immunity and Infection (Formerly Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
40
|
Jeong K, Chang J, Park SM, Kim J, Jeon S, Kim DH, Kim YE, Lee JC, Im S, Jo Y, Min JY, Lee H, Yeom M, Seok SH, On DI, Noh H, Yun JW, Park JW, Song D, Seong JK, Kim KC, Lee JY, Park HJ, Kim S, Nam TG, Lee W. Rapid discovery and classification of inhibitors of coronavirus infection by pseudovirus screen and amplified luminescence proximity homogeneous assay. Antiviral Res 2023; 209:105473. [PMID: 36435212 PMCID: PMC9682871 DOI: 10.1016/j.antiviral.2022.105473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022]
Abstract
To identify potent antiviral compounds, we introduced a high-throughput screen platform that can rapidly classify hit compounds according to their target. In our platform, we performed a compound screen using a lentivirus-based pseudovirus presenting a spike protein of coronavirus, and we evaluated the hit compounds using an amplified luminescence proximity homogeneous assay (alpha) test with purified host receptor protein and the receptor binding domain of the viral spike. With our screen platform, we were able to identify both spike-specific compounds (class I) and broad-spectrum antiviral compounds (class II). Among the hit compounds, thiosemicarbazide was identified to be selective to the interaction between the viral spike and its host cell receptor, and we further optimized the binding potency of thiosemicarbazide through modification of the pyridine group. Among the class II compounds, we found raloxifene and amiodarone to be highly potent against human coronaviruses including Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus (SARS-CoV), and SARS-CoV-2. In particular, using analogs of the benzothiophene moiety, which is also present in raloxifene, we have identified benzothiophene as a novel structural scaffold for broad-spectrum antivirals. This work highlights the strong utility of our screen platform using a pseudovirus assay and an alpha test for rapid identification of potential antiviral compounds and their mechanism of action, which can lead to the accelerated development of therapeutics against newly emerging viral infections.
Collapse
Affiliation(s)
- Kwiwan Jeong
- Bio-center, Gyeonggido Business and Science Accelerator, Suwon, South Korea,Corresponding author
| | - JuOae Chang
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sun-mi Park
- Bio-center, Gyeonggido Business and Science Accelerator, Suwon, South Korea
| | - Jinhee Kim
- Institut Pasteur Korea, Seongnam, South Korea
| | | | - Dong Hwan Kim
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Young-Eui Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Joo Chan Lee
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Somyoung Im
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Yejin Jo
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | | | - Hanbyeul Lee
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Minjoo Yeom
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Sang-Hyuk Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, South Korea
| | - Da In On
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Hyuna Noh
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, South Korea
| | - Daesub Song
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea,Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, South Korea
| | - Kyung-Chang Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Joo-Yeon Lee
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Hyun-Ju Park
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea,Corresponding author
| | - Seungtaek Kim
- Institut Pasteur Korea, Seongnam, South Korea,Corresponding author
| | - Tae-gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea,Corresponding author
| | - Wonsik Lee
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea,Corresponding author
| |
Collapse
|
41
|
Guo C, Tsai SJ, Ai Y, Li M, Anaya E, Pekosz A, Cox A, Gould SJ. The D614G mutation redirects SARS-CoV-2 spike to lysosomes and suppresses deleterious traits of the furin cleavage site insertion mutation. SCIENCE ADVANCES 2022; 8:eade5085. [PMID: 36563151 PMCID: PMC9788772 DOI: 10.1126/sciadv.ade5085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/21/2022] [Indexed: 06/16/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) egress occurs by lysosomal exocytosis. We show that the Spike D614G mutation enhances Spike trafficking to lysosomes, drives Spike-mediated reprogramming of lysosomes, and reduces cell surface Spike expression by ~3-fold. D614G is not a human-specific adaptation. Rather, it is an adaptation to the earlier furin cleavage site insertion (FCSI) mutation that occurred at the genesis of SARS-CoV-2. While advantageous to the virus, furin cleavage of spike has deleterious effects on spike structure and function, inhibiting its trafficking to lysosomes and impairing its infectivity by the transmembrane serine protease 2(TMPRSS2)-independent, endolysosomal pathway. D614G restores spike trafficking to lysosomes and enhances the earliest events in SARS-CoV-2 infectivity, while spike mutations that restore SARS-CoV-2's TMPRSS2-independent infectivity restore spike's trafficking to lysosomes. Together, these and other results show that D614G is an intragenic suppressor of deleterious traits linked to the FCSI and lend additional support to the endolysosomal model of SARS-CoV-2 egress and entry.
Collapse
Affiliation(s)
- Chenxu Guo
- Department of Biological Chemistry, Johns Hopkins University, School of Medicine, 725 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Shang-Jui Tsai
- Department of Biological Chemistry, Johns Hopkins University, School of Medicine, 725 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Yiwei Ai
- Department of Biological Chemistry, Johns Hopkins University, School of Medicine, 725 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Maggie Li
- Department of Microbiology and Immunology, Johns Hopkins University, School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Eduardo Anaya
- Department of Microbiology and Immunology, Johns Hopkins University, School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Andrew Pekosz
- Department of Microbiology and Immunology, Johns Hopkins University, School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Andrea Cox
- Department of Medicine, Department of Microbiology and Immunology, Johns Hopkins University, School of Medicine, 725 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Stephen J. Gould
- Department of Biological Chemistry, Johns Hopkins University, School of Medicine, 725 North Wolfe Street, Baltimore, MD, 21205, USA
| |
Collapse
|
42
|
Stewart H, Palmulli R, Johansen KH, McGovern N, Shehata OM, Carnell GW, Jackson HK, Lee JS, Brown JC, Burgoyne T, Heeney JL, Okkenhaug K, Firth AE, Peden AA, Edgar JR. Tetherin antagonism by SARS-CoV-2 enhances virus release: multiple mechanisms including ORF3a-mediated defective retrograde traffic. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.01.06.425396. [PMID: 33442692 PMCID: PMC7805449 DOI: 10.1101/2021.01.06.425396] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The antiviral restriction factor, tetherin, blocks the release of several different families of enveloped viruses, including the Coronaviridae. Tetherin is an interferon-induced protein that forms parallel homodimers between the host cell and viral particles, linking viruses to the surface of infected cells and inhibiting their release. We demonstrated that SARS-CoV-2 infection causes tetherin downregulation, and that tetherin depletion from cells enhances SARS-CoV-2 viral titres. We investigated the potential viral proteins involved in abrogating tetherin function and found that SARS-CoV-2 ORF3a reduces tetherin localisation within biosynthetic organelles via reduced retrograde recycling and increases tetherin localisation to late endocytic organelles. By removing tetherin from the Coronavirus budding compartments, ORF3a enhances virus release. We also found expression of Spike protein caused a reduction in cellular tetherin levels. Our results confirm that tetherin acts as a host restriction factor for SARS-CoV-2 and highlight the multiple distinct mechanisms by which SARS-CoV-2 subverts tetherin function.
Collapse
Affiliation(s)
- Hazel Stewart
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
| | - Roberta Palmulli
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
| | - Kristoffer H. Johansen
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
- Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
| | - Ola M. Shehata
- Department of Biomedical Science, University of Sheffield, Firth Court, Sheffield. S10 2TN. UK
| | - George W. Carnell
- Department of Veterinary Medicine, University of Cambridge, Cambridge. CB3 0ES. UK
| | - Hannah K. Jackson
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
| | - Jin S. Lee
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
| | - Jonathan C. Brown
- Department of Infectious Disease, Imperial College London, London. W2 1PG. UK
| | - Thomas Burgoyne
- Royal Brompton Hospital, Guy’s and St Thomas’ NHS Foundation Trust, London. SW3 6NP. UK
- UCL Institute of Ophthalmology, University College London, London. EC1V 9EL. UK
| | - Jonathan L. Heeney
- Department of Veterinary Medicine, University of Cambridge, Cambridge. CB3 0ES. UK
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
| | - Andrew E. Firth
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
| | - Andrew A. Peden
- Department of Biomedical Science, University of Sheffield, Firth Court, Sheffield. S10 2TN. UK
| | - James R. Edgar
- Department of Pathology, University of Cambridge, Cambridge. CB2 1QP. UK
| |
Collapse
|
43
|
Matveeva O, Nechipurenko Y, Lagutkin D, Yegorov YE, Kzhyshkowska J. SARS-CoV-2 infection of phagocytic immune cells and COVID-19 pathology: Antibody-dependent as well as independent cell entry. Front Immunol 2022; 13:1050478. [PMID: 36532011 PMCID: PMC9751203 DOI: 10.3389/fimmu.2022.1050478] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
Our review summarizes the evidence that COVID-19 can be complicated by SARS-CoV-2 infection of immune cells. This evidence is widespread and accumulating at an increasing rate. Research teams from around the world, studying primary and established cell cultures, animal models, and analyzing autopsy material from COVID-19 deceased patients, are seeing the same thing, namely that some immune cells are infected or capable of being infected with the virus. Human cells most vulnerable to infection include both professional phagocytes, such as monocytes, macrophages, and dendritic cells, as well as nonprofessional phagocytes, such as B-cells. Convincing evidence has accumulated to suggest that the virus can infect monocytes and macrophages, while data on infection of dendritic cells and B-cells are still scarce. Viral infection of immune cells can occur directly through cell receptors, but it can also be mediated or enhanced by antibodies through the Fc gamma receptors of phagocytic cells. Antibody-dependent enhancement (ADE) most likely occurs during the primary encounter with the pathogen through the first COVID-19 infection rather than during the second encounter, which is characteristic of ADE caused by other viruses. Highly fucosylated antibodies of vaccinees seems to be incapable of causing ADE, whereas afucosylated antibodies of persons with acute primary infection or convalescents are capable. SARS-CoV-2 entry into immune cells can lead to an abortive infection followed by host cell pyroptosis, and a massive inflammatory cascade. This scenario has the most experimental evidence. Other scenarios are also possible, for which the evidence base is not yet as extensive, namely productive infection of immune cells or trans-infection of other non-immune permissive cells. The chance of a latent infection cannot be ruled out either.
Collapse
Affiliation(s)
- Olga Matveeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Denis Lagutkin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Center of Phthisiopulmonology and Infectious Diseases under the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yegor E. Yegorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- German Red Cross Blood Service Baden-Württemberg – Hessen, Mannheim, Germany
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| |
Collapse
|
44
|
Borin A, Coimbra LD, Bispo-dos-Santos K, Naciuk FF, Fontoura M, Simeoni CL, Gomes GV, Amorim MR, Gravina HD, Shimizu JF, Passos ASC, de Oliveira IM, de Carvalho AC, Cardoso AC, Parise PL, Toledo-Teixeira DA, Sotorilli GE, Persinoti GF, Claro IM, Sabino EC, Alborghetti MR, Rocco SA, Franchini KG, de Souza WM, Oliveira PSL, Cunha TM, Granja F, Proença-Módena JL, Trivella DB, Bruder M, Cordeiro AT, Marques RE. Identification and characterization of the anti-SARS-CoV-2 activity of cationic amphiphilic steroidal compounds. Virulence 2022; 13:1031-1048. [PMID: 35734825 PMCID: PMC9235892 DOI: 10.1080/21505594.2022.2085793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/13/2022] [Accepted: 05/31/2022] [Indexed: 11/11/2022] Open
Abstract
The ongoing COVID-19 pandemic caused a significant loss of human lives and a worldwide decline in quality of life. Treatment of COVID-19 patients is challenging, and specific treatments to reduce COVID-19 aggravation and mortality are still necessary. Here, we describe the discovery of a novel class of epiandrosterone steroidal compounds with cationic amphiphilic properties that present antiviral activity against SARS-CoV-2 in the low micromolar range. Compounds were identified in screening campaigns using a cytopathic effect-based assay in Vero CCL81 cells, followed by hit compound validation and characterization. Compounds LNB167 and LNB169 were selected due to their ability to reduce the levels of infectious viral progeny and viral RNA levels in Vero CCL81, HEK293, and HuH7.5 cell lines. Mechanistic studies in Vero CCL81 cells indicated that LNB167 and LNB169 inhibited the initial phase of viral replication through mechanisms involving modulation of membrane lipids and cholesterol in host cells. Selection of viral variants resistant to steroidal compound treatment revealed single mutations on transmembrane, lipid membrane-interacting Spike and Envelope proteins. Finally, in vivo testing using the hACE2 transgenic mouse model indicated that SARS-CoV-2 infection could not be ameliorated by LNB167 treatment. We conclude that anti-SARS-CoV-2 activities of steroidal compounds LNB167 and LNB169 are likely host-targeted, consistent with the properties of cationic amphiphilic compounds that modulate host cell lipid biology. Although effective in vitro, protective effects were cell-type specific and did not translate to protection in vivo, indicating that subversion of lipid membrane physiology is an important, yet complex mechanism involved in SARS-CoV-2 replication and pathogenesis.
Collapse
Affiliation(s)
- Alexandre Borin
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Laís D. Coimbra
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Karina Bispo-dos-Santos
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Fabrício F. Naciuk
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Marina Fontoura
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
- Department of Cellular and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Camila L. Simeoni
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Giovanni V. Gomes
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Mariene R. Amorim
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Humberto D. Gravina
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Jacqueline Farinha Shimizu
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Amanda S. C. Passos
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Isadora M. de Oliveira
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Ana Carolina de Carvalho
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Alisson Campos Cardoso
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Pierina L. Parise
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniel A. Toledo-Teixeira
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Giuliana E. Sotorilli
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Gabriela F. Persinoti
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), São Paulo, Brazil
| | - Ingra Morales Claro
- Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Ester C. Sabino
- Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Marcos R. Alborghetti
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Silvana A. Rocco
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Kleber G. Franchini
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - William M. de Souza
- World Reference Center for Emerging Viruses and Arboviruses and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Paulo S. L. Oliveira
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Thiago M. Cunha
- Center for Research in Inflammatory Diseases (CRID), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fabiana Granja
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Biodiversity Research Center, Federal University of Roraima (UFRR), Boa Vista, Brazil
| | - José Luiz Proença-Módena
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
- Experimental Medicine Research Cluster, University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniela B.B. Trivella
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Marjorie Bruder
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Artur T. Cordeiro
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory - LNBio, Brazilian Center for Research in Energy and Materials - CNPEM, Campinas, Brazil
| |
Collapse
|
45
|
Mao B, Le-Trilling VTK, Wang K, Mennerich D, Hu J, Zhao Z, Zheng J, Deng Y, Katschinski B, Xu S, Zhang G, Cai X, Hu Y, Wang J, Lu M, Huang A, Tang N, Trilling M, Lin Y. Obatoclax inhibits SARS-CoV-2 entry by altered endosomal acidification and impaired cathepsin and furin activity in vitro. Emerg Microbes Infect 2022; 11:483-497. [PMID: 34989664 PMCID: PMC8843317 DOI: 10.1080/22221751.2022.2026739] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 12/25/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has set off a global pandemic. There is an urgent unmet need for safe, affordable, and effective therapeutics against COVID-19. In this regard, drug repurposing is considered as a promising approach. We assessed the compounds that affect the endosomal acidic environment by applying human angiotensin-converting enzyme 2 (hACE2)- expressing cells infected with a SARS-CoV-2 spike (S) protein-pseudotyped HIV reporter virus and identified that obatoclax resulted in the strongest inhibition of S protein-mediated virus entry. The potent antiviral activity of obatoclax at nanomolar concentrations was confirmed in different human lung and intestinal cells infected with the SARS-CoV-2 pseudotype system as well as clinical virus isolates. Furthermore, we uncovered that obatoclax executes a double-strike against SARS-CoV-2. It prevented SARS-CoV-2 entry by blocking endocytosis of virions through diminished endosomal acidification and the corresponding inhibition of the enzymatic activity of the endosomal cysteine protease cathepsin L. Additionally, obatoclax impaired the SARS-CoV-2 S-mediated membrane fusion by targeting the MCL-1 protein and reducing furin protease activity. In accordance with these overarching mechanisms, obatoclax blocked the virus entry mediated by different S proteins derived from several SARS-CoV-2 variants of concern such as, Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2). Taken together, our results identified obatoclax as a novel effective antiviral compound that keeps SARS-CoV-2 at bay by blocking both endocytosis and membrane fusion. Our data suggested that obatoclax should be further explored as a clinical drug for the treatment of COVID-19.
Collapse
Affiliation(s)
- Binli Mao
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | | | - Kai Wang
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Denise Mennerich
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jie Hu
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Zhenyu Zhao
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jiaxin Zheng
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yingying Deng
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Benjamin Katschinski
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Shilei Xu
- Department of General Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Guiji Zhang
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xuefei Cai
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yuan Hu
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jianwei Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ailong Huang
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ni Tang
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Yong Lin
- Key Laboratory of Molecular Biology of Infectious Diseases (Chinese Ministry of Education), Department of Infectious Diseases, The Second Affiliated Hospital, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
46
|
Ruiz MJ, Siracusano G, Cottignies-Calamarte A, Tudor D, Real F, Zhu A, Pastori C, Capron C, Rosenberg AR, Temperton N, Cantoni D, Liao H, Ternette N, Moine P, Godement M, Geri G, Chiche JD, Annane D, Cramer Bordé E, Lopalco L, Bomsel M. Persistent but dysfunctional mucosal SARS-CoV-2-specific IgA and low lung IL-1β associate with COVID-19 fatal outcome: A cross-sectional analysis. Front Immunol 2022; 13:842468. [PMID: 36248831 PMCID: PMC9560774 DOI: 10.3389/fimmu.2022.842468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
The role of the mucosal pulmonary antibody response in coronavirus disease 2019 (COVID-19) outcome remains unclear. Here, we found that in bronchoalveolar lavage (BAL) samples from 48 patients with severe COVID-19-infected with the ancestral Wuhan virus, mucosal IgG and IgA specific for S1, receptor-binding domain (RBD), S2, and nucleocapsid protein (NP) emerged in BAL containing viruses early in infection and persist after virus elimination, with more IgA than IgG for all antigens tested. Furthermore, spike-IgA and spike-IgG immune complexes were detected in BAL, especially when the lung virus has been cleared. BAL IgG and IgA recognized the four main RBD variants. BAL neutralizing titers were higher early in COVID-19 when virus replicates in the lung than later in infection after viral clearance. Patients with fatal COVID-19, in contrast to survivors, developed higher levels of mucosal spike-specific IgA than IgG but lost neutralizing activities over time and had reduced IL-1β in the lung. Altogether, mucosal spike and NP-specific IgG and S1-specific IgA persisting after lung severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) clearance and low pulmonary IL-1β correlate with COVID-19 fatal outcome. Thus, mucosal SARS-CoV-2-specific antibodies may have adverse functions in addition to protective neutralization.
Collapse
Affiliation(s)
- Maria Julia Ruiz
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Gabriel Siracusano
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Andréa Cottignies-Calamarte
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Daniela Tudor
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Fernando Real
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Aiwei Zhu
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Claudia Pastori
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Claude Capron
- AP-HP, Hôpital Ambroise Paré, Service d'Hématologie, Boulogne-Billancourt, France
| | - Arielle R. Rosenberg
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- AP-HP, Hôpital Cochin, Service de Virologie, Paris, France
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Hanqing Liao
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicola Ternette
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Pierre Moine
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Mathieu Godement
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Guillaume Geri
- AP-HP, Hôpital Ambroise Paré, Service de Réanimation, Boulogne-Billancourt, France
- Université de Versailles-St Quentin en Yvelines, Versailles, France
| | | | - Djillali Annane
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | | | - Lucia Lopalco
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Morgane Bomsel
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- *Correspondence: Morgane Bomsel,
| |
Collapse
|
47
|
Hou W, Wang S, Wu H, Xue L, Wang B, Wang S, Wang H. Small GTPase-a Key Role in Host Cell for Coronavirus Infection and a Potential Target for Coronavirus Vaccine Adjuvant Discovery. Viruses 2022; 14:v14092044. [PMID: 36146850 PMCID: PMC9504349 DOI: 10.3390/v14092044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/20/2022] Open
Abstract
Small GTPases are signaling molecules in regulating key cellular processes (e.g., cell differentiation, proliferation, and motility) as well as subcellular events (e.g., vesicle trafficking), making them key participants, especially in a great array of coronavirus infection processes. In this review, we discuss the role of small GTPases in the coronavirus life cycle, especially pre-entry, endocytosis, intracellular traffic, replication, and egress from the host cell. Furthermore, we also suggest the molecules that have potent adjuvant activity by targeting small GTPases. These studies provide deep insights and references to understand the pathogenesis of coronavirus as well as to propose the potential of small GTPases as targets for adjuvant development.
Collapse
Affiliation(s)
- Wei Hou
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Sibei Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Heqiong Wu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Linli Xue
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Bin Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
- Single Molecule Nanometry Laboratory (Sinmolab), Nanjing Agricultural University, Nanjing 210095, China
| | | | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
- Correspondence:
| |
Collapse
|
48
|
Mahata P, Vennamneni L, Chattopadhyay S. A mechanical-thermodynamic model for understanding endocytosis of COVID-19 virus SARS-CoV-2. PROCEEDINGS OF THE INSTITUTION OF MECHANICAL ENGINEERS. PART C. JOURNAL OF MECHANICAL ENGINEERING SCIENCE 2022; 236:9431-9440. [PMID: 38603131 PMCID: PMC9127454 DOI: 10.1177/09544062221098538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 04/11/2022] [Indexed: 04/13/2024]
Abstract
We analyze the endocytosis process of COVID-19 (coronavirus disease 2019) virus SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) using a mechanical-thermodynamic model. The virus particle is designed to interface with the cell membrane as a hard sphere. The role of cytoplasmic BAR (Bin/Amphiphysin/RVs) proteins is considered in the endocytosis. Interestingly, the Endophilin N-BAR cytoplasmic proteins show resistance in participating endocytosis, whereas F-BAR, Arfaptin BAR, Amphiphysin N-BAR, and PX-BAR proteins participate in endocytosis. The increase in membrane tension, concentrated force between the cell membrane receptor, and spike glycoprotein present on the surface of virus particle promote the endocytosis. Also, the increase in the bending modulus of membrane leads to the two-phase solution of BAR protein concentration on the interior of cell membrane surface. We observe an unstable region of protein concentration, which may help one to retard the endocytosis process and thus the viral infection. Though the present study is focused on SARS-CoV-2, it can be extended to understand any other viral infections, involving endocytosis process.
Collapse
Affiliation(s)
- Paritosh Mahata
- Department of Mechanical Engineering, Birla Institute of Technology, Ranchi, India
| | | | | |
Collapse
|
49
|
Yan Q, Wu K, Zeng W, Yu S, Li Y, Sun Y, Liu X, Ruan Y, Huang J, Ding H, Yi L, Zhao M, Chen J, Fan S. Historical Evolutionary Dynamics and Phylogeography Analysis of Transmissible Gastroenteritis Virus and Porcine Deltacoronavirus: Findings from 59 Suspected Swine Viral Samples from China. Int J Mol Sci 2022; 23:ijms23179786. [PMID: 36077190 PMCID: PMC9456201 DOI: 10.3390/ijms23179786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/14/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Since the beginning of the 21st century, humans have experienced three coronavirus pandemics, all of which were transmitted to humans via animals. Recent studies have found that porcine deltacoronavirus (PDCoV) can infect humans, so swine enteric coronavirus (SeCoV) may cause harm through cross-species transmission. Transmissible gastroenteritis virus (TGEV) and PDCoV have caused tremendous damage and loss to the pig industry around the world. Therefore, we analyzed the genome sequence data of these two SeCoVs by evolutionary dynamics and phylogeography, revealing the genetic diversity and spatiotemporal distribution characteristics. Maximum likelihood and Bayesian inference analysis showed that TGEV could be divided into two different genotypes, and PDCoV could be divided into four main lineages. Based on the analysis results inferred by phylogeography, we inferred that TGEV might originate from America, PDCoV might originate from Asia, and different migration events had different migration rates. In addition, we also identified positive selection sites of spike protein in TGEV and PDCoV, indicating that the above sites play an essential role in promoting membrane fusion to achieve adaptive evolution. In a word, TGEV and PDCoV are the past and future of SeCoV, and the relatively smooth transmission rate of TGEV and the increasing transmission events of PDCoV are their respective transmission characteristics. Our results provide new insights into the evolutionary characteristics and transmission diversity of these SeCoVs, highlighting the potential for cross-species transmission of SeCoV and the importance of enhanced surveillance and biosecurity measures for SeCoV in the context of the COVID-19 epidemic.
Collapse
Affiliation(s)
- Quanhui Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Keke Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Weijun Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Shu Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yuwan Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yawei Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Xiaodi Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yang Ruan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Juncong Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: (J.C.); (S.F.); Tel.: +86-20-8528-8017 (J.C.); +86-20-8528-8017 (S.F.)
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: (J.C.); (S.F.); Tel.: +86-20-8528-8017 (J.C.); +86-20-8528-8017 (S.F.)
| |
Collapse
|
50
|
Tanaka Y, Tanabe E, Nonaka Y, Uemura M, Tajima T, Ochiai K. Ionophore Antibiotics Inhibit Type II Feline Coronavirus Proliferation In Vitro. Viruses 2022; 14:v14081734. [PMID: 36016355 PMCID: PMC9415497 DOI: 10.3390/v14081734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 12/03/2022] Open
Abstract
Feline coronaviruses (FCoVs) infect cats worldwide and cause severe systemic diseases, such as feline infectious peritonitis (FIP). FIP has a high mortality rate, and drugs approved by the Food and Drug Administration have been ineffective for the treatment of FIP. Investigating host factors and the functions required for FCoV replication is necessary to develop effective drugs for the treatment of FIP. FCoV utilizes an endosomal trafficking system for cellular entry after binding between the viral spike (S) protein and its receptor. The cellular enzymes that cleave the S protein of FCoV to release the viral genome into the cytosol require an acidic pH optimized in the endosomes by regulating cellular ion concentrations. Ionophore antibiotics are compounds that form complexes with alkali ions to alter the endosomal pH conditions. This study shows that ionophore antibiotics, including valinomycin, salinomycin, and nigericin, inhibit FCoV proliferation in vitro in a dose-dependent manner. These results suggest that ionophore antibiotics should be investigated further as potential broad-spectrum anti-FCoV agents.
Collapse
Affiliation(s)
- Yoshikazu Tanaka
- Department of Veterinary Hygiene, Veterinary School, Nippon Veterinary & Life Science University, 1-7-1 Kyounan, Musashino 180-8602, Japan
- Research Center for Animal Life Science, Nippon Veterinary & Life Science University, 1-7-1 Kyounan, Musashino 180-8602, Japan
- Correspondence: ; Tel.: +81-422-31-4151
| | - Eri Tanabe
- Department of Veterinary Hygiene, Veterinary School, Nippon Veterinary & Life Science University, 1-7-1 Kyounan, Musashino 180-8602, Japan
| | - Yuki Nonaka
- Department of Veterinary Hygiene, Veterinary School, Nippon Veterinary & Life Science University, 1-7-1 Kyounan, Musashino 180-8602, Japan
| | - Mitsuki Uemura
- Department of Veterinary Hygiene, Veterinary School, Nippon Veterinary & Life Science University, 1-7-1 Kyounan, Musashino 180-8602, Japan
| | - Tsuyoshi Tajima
- Department of Veterinary Pharmacology, Veterinary School, Nippon Veterinary & Life Science University, 1-7-1 Kyounan, Musashino 180-8602, Japan
| | - Kazuhiko Ochiai
- Department of Veterinary Hygiene, Veterinary School, Nippon Veterinary & Life Science University, 1-7-1 Kyounan, Musashino 180-8602, Japan
- Research Center for Animal Life Science, Nippon Veterinary & Life Science University, 1-7-1 Kyounan, Musashino 180-8602, Japan
| |
Collapse
|