1
|
Nowak R, Gazecka M, Hoffmann M, Kierzek R, Pöhlmann S, Zmora P. TMPRSS2-specific antisense oligonucleotides inhibit host cell entry of emerging viruses. Virology 2024; 600:110218. [PMID: 39276670 DOI: 10.1016/j.virol.2024.110218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/06/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024]
Abstract
Emerging viruses, such as novel influenza A viruses (IAV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), pose a constant threat to animal and human health. Identification of host cell factors necessary for viral replication but dispensable for cellular survival might reveal novel, attractive targets for therapeutic intervention. Proteolytic activation of IAV hemagglutinin (HA) and SARS-CoV-2 spike protein (S) by the type II transmembrane serine protease (TTSPs), e.g. TMPRSS2 is sought to be critical for viral spread and pathogenesis. Here, we investigated the secondary structure of TMPRSS2 mRNA coding sequence and designed TMPRSS2-specific antisense oligonucleotides (ASOs). Several of these ASOs markedly reduced the TMPRSS2 expression and decreased IAV infection and SARS-CoV-2 entry into cells.
Collapse
Affiliation(s)
- Rafal Nowak
- Department of Molecular Virology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Monika Gazecka
- Department of Molecular Virology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - 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
| | - Ryszard Kierzek
- Department of Structural Chemistry and Biology of Nucleic Acids, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany; Faculty of Biology and Psychology, Georg August University, Göttingen, Germany
| | - Pawel Zmora
- Department of Molecular Virology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland.
| |
Collapse
|
2
|
Houghton MJ, Balland E, Gartner MJ, Thomas BJ, Subbarao K, Williamson G. The flavonoid quercetin decreases ACE2 and TMPRSS2 expression but not SARS-CoV-2 infection in cultured human lung cells. Biofactors 2024; 50:1268-1286. [PMID: 38886986 PMCID: PMC11627474 DOI: 10.1002/biof.2084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/11/2024] [Indexed: 06/20/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binds to angiotensin-converting enzyme 2 (ACE2) on host cells, via its spike protein, and transmembrane protease, serine 2 (TMPRSS2) cleaves the spike-ACE2 complex to facilitate virus entry. As rate-limiting steps for virus entry, modulation of ACE2 and/or TMPRSS2 may decrease SARS-CoV-2 infectivity and COVID-19 severity. In silico modeling suggested the natural bioactive flavonoid quercetin can bind to ACE2 and a recent randomized clinical trial demonstrated that oral supplementation with quercetin increased COVID-19 recovery. A range of cultured human cells were assessed for co-expression of ACE2 and TMPRSS2. Immortalized Calu-3 lung cells, cultured and matured at an air-liquid interface (Calu-3-ALIs), were established as the most appropriate. Primary bronchial epithelial cells (PBECs) were obtained from healthy adult males (N = 6) and cultured under submerged conditions to corroborate the outcomes. Upon maturation or reaching 80% confluence, respectively, the Calu-3-ALIs and PBECs were treated with quercetin, and mRNA and protein expression were assessed by droplet digital PCR and ELISA, respectively. SARS-CoV-2 infectivity, and the effects of pre- and co-treatment with quercetin, was assessed by median tissue culture infectious dose assay. Quercetin dose-dependently decreased ACE2 and TMPRSS2 mRNA and protein in both Calu-3-ALIs and PBECs after 4 h, while TMPRSS2 remained suppressed in response to prolonged treatment with lower doses (twice daily for 3 days). Quercetin also acutely decreased ADAM17 mRNA, but not ACE, in Calu-3-ALIs, and this warrants further investigation. Calu-3-ALIs, but not PBECs, were successfully infected with SARS-CoV-2; however, quercetin had no antiviral effect, neither directly nor indirectly through downregulation of ACE2 and TMPRSS2. Calu-3-ALIs were reaffirmed to be an optimal cell model for research into the regulation of ACE2 and TMPRSS2, without the need for prior genetic modification, and will prove valuable in future coronavirus and respiratory infectious disease work. However, our data demonstrate that a significant decrease in the expression of ACE2 and TMPRSS2 by a promising prophylactic candidate may not translate to infection prevention.
Collapse
Affiliation(s)
- Michael James Houghton
- Department of Nutrition, Dietetics and FoodMonash University, BASE FacilityNotting HillVICAustralia
- Victorian Heart InstituteMonash University, Victorian Heart HospitalClaytonVICAustralia
| | - Eglantine Balland
- Department of Nutrition, Dietetics and FoodMonash University, BASE FacilityNotting HillVICAustralia
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental BiologyMonash UniversityClaytonVICAustralia
| | - Matthew James Gartner
- Department of Microbiology and ImmunologyUniversity of Melbourne at The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Belinda Jane Thomas
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Monash Lung and Sleep, Monash Health, Monash Medical CentreClaytonVICAustralia
| | - Kanta Subbarao
- Department of Microbiology and ImmunologyUniversity of Melbourne at The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
- WHO Collaborating Centre for Reference and Research on InfluenzaThe Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Gary Williamson
- Department of Nutrition, Dietetics and FoodMonash University, BASE FacilityNotting HillVICAustralia
- Victorian Heart InstituteMonash University, Victorian Heart HospitalClaytonVICAustralia
| |
Collapse
|
3
|
Calcagnile M, Damiano F, Lobreglio G, Siculella L, Bozzetti MP, Forgez P, Malgoyre A, Libert N, Bucci C, Alifano M, Alifano P. In silico evidence that substitution of glycine for valine (p.G8V) in a common variant of TMPRSS2 isoform 1 increases accessibility to an endocytic signal: Implication for SARS-cov-2 entry into host cells and susceptibility to COVID-19. Biochimie 2024; 225:89-98. [PMID: 38754620 DOI: 10.1016/j.biochi.2024.05.004] [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: 02/16/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024]
Abstract
The TMPRSS2 protease plays a key role in the entry of the SARS-CoV-2 into cells. The TMPRSS2 gene is highly polymorphic in humans, and some polymorphisms may affect the susceptibility to COVID-19 or disease severity. rs75603675 (c.23G > T) is a missense variant that causes the replacement of glycine with valine at position 8 (p.G8V) in the TMPRSS2 isoform 1. According to GnomAD v4.0.0 database, the allele frequency of the rs75603675 on a global scale is 38.10 %, and range from 0.92 % in East Asian to 40.77 % in non-Finnish European (NFE) population. We analyzed the occurrence of the rs75603675 in two cohorts of patients, the first with severe/critical COVID-19 enrolled in a French hospital (42 patients), and the second with predominantly asymptomatic/pauci-symptomatic/mild COVID-19 enrolled in an Italian hospital (69 patients). We found that the TMPRSS2-c.23T minor allele frequency was similar in the two cohorts, 46.43 % and 46.38 %, respectively, and higher than the frequency in the NFE population (40.77 %). Chi-square test provided significant results (p < 0.05) when the genotype data (TMPRSS2-c.23T/c.23T homozygotes + TMPRSS2-c.23G/c.23T heterozygotes vs. TMPRSS2-c.23G/c.23G homozygotes) of the two patient groups were pooled and compared to the expected data for the NFE population, suggesting a possible pathogenetic mechanism of the p.G8V substitution. We explored the possible effects of the p.G8V substitution and found that the N-terminal region of the TMPRSS2 isoform 1 contains a signal for clathrin/AP-2-dependent endocytosis. In silico analysis predicted that the p.G8V substitution may increase the accessibility to the endocytic signal, which could help SARS-CoV-2 enter cells.
Collapse
Affiliation(s)
- Matteo Calcagnile
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Fabrizio Damiano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Giambattista Lobreglio
- Clinical Pathology and Microbiology Unit, Vito Fazzi General Hospital, 73100, Lecce, Italy
| | - Luisa Siculella
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Maria Pia Bozzetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Patricia Forgez
- INSERM UMR-S 1124 T3S, Eq 5 CELLULAR HOMEOSTASIS, CANCER and THERAPY, University of Paris, Campus Saint Germain, Paris, France
| | - Alexandra Malgoyre
- Institut de Recherche Biomedicale des Armées, French Armed Forces Health Services, Brétigny sur Orge, France; Ecole Du Val de Grâce, French Armed Forces Health Service, France; Laboratoire de Biologie de L'Exercice pour La Performance et La Santé, Université Evry-Paris-Saclay, Evry, France
| | - Nicolas Libert
- Ecole Du Val de Grâce, French Armed Forces Health Service, France; Hopital D'Instruction des Armées, French Armed Forces Health Services, Clamart, France
| | - Cecilia Bucci
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, France; INSERM U1138 Team «Cancer, Immune Control, and Escape», Cordeliers Research Center, University of Paris, France.
| | - Pietro Alifano
- Department of Experimental Medicine, University of Salento, Lecce, Italy.
| |
Collapse
|
4
|
Sharanya CS, Wilbee DS, Sathi SN, Natarajan K. Computational screening combined with well-tempered metadynamics simulations identifies potential TMPRSS2 inhibitors. Sci Rep 2024; 14:16197. [PMID: 39003338 PMCID: PMC11246518 DOI: 10.1038/s41598-024-65296-7] [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: 05/10/2024] [Accepted: 06/19/2024] [Indexed: 07/15/2024] Open
Abstract
Type-II transmembrane serine proteases are effective pharmacological targets for host defence against viral entry and in certain cancer cell progressions. These serine proteases cleave viral spike proteins to expose the fusion peptide for cell entry, which is essential to the life cycle of the virus. TMPRSS2 inhibitors can also fight against respiratory viruses that employ them for cell entry. Our study combining virtual screening, all-atom molecular dynamics, and well-tempered metadynamics simulation identifies vicenin-2, neohesperidin, naringin, and rhoifolin as promising TMPRSS2 antagonists. The binding energies obtained are - 16.3, - 15.4, - 13.6, and - 13.8 kcal/mol for vicenin-2, neohesperidin, naringin, and rhoifolin respectively. The RMSD, RMSF, PCA, DCCM, and binding free energy profiles also correlate with the stable binding of these ligands at the active site of TMPRSS2. The study reveals that these molecules could be promising lead molecules for combating future outbreaks of coronavirus and other respiratory viruses.
Collapse
Affiliation(s)
- C S Sharanya
- Transdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - D Sasikala Wilbee
- Transdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
- College of Pharmaceutical Sciences, Government Medical College, Thiruvananthapuram, Kerala, India
| | - Shijulal Nelson Sathi
- Bioinformatics Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Kathiresan Natarajan
- Transdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
5
|
De Magis A, Schult P, Schönleber A, Linke R, Ludwig KU, Kümmerer BM, Paeschke K. TMPRSS2 isoform 1 downregulation by G-quadruplex stabilization induces SARS-CoV-2 replication arrest. BMC Biol 2024; 22:5. [PMID: 38185627 PMCID: PMC10773119 DOI: 10.1186/s12915-023-01805-w] [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: 03/02/2023] [Accepted: 12/18/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND SARS-CoV-2 infection depends on the host cell factors angiotensin-converting enzyme 2, ACE2, and the transmembrane serinprotease 2, TMPRSS2. Potential inhibitors of these proteins would be ideal targets against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection. Our data opens the possibility that changes within TMPRSS2 can modulate the outcome during a SARS-CoV-2 infection. RESULTS We reveal that TMPRSS2 acts not only during viral entry but has also an important role during viral replication. In addition to previous functions for TMPRSS2 during viral entry, we determined by specific downregulation of distinct isoforms that only isoform 1 controls and supports viral replication. G-quadruplex (G4) stabilization by chemical compounds impacts TMPRSS2 gene expression. Here we extend and in-depth characterize these observations and identify that a specific G4 in the first exon of the TMPRSS2 isoform 1 is particular targeted by the G4 ligand and affects viral replication. Analysis of potential single nucleotide polymorphisms (SNPs) reveals that a reported SNP at this G4 in isoform 1 destroys the G4 motif and makes TMPRSS2 ineffective towards G4 treatment. CONCLUSION These findings uncover a novel mechanism in which G4 stabilization impacts SARS-CoV-2 replication by changing TMPRSS2 isoform 1 gene expression.
Collapse
Affiliation(s)
- Alessio De Magis
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- Department of Oncology, Haematology and Rheumatology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Philipp Schult
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Oncology, Haematology and Rheumatology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Antonia Schönleber
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Rebecca Linke
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Oncology, Haematology and Rheumatology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Kerstin U Ludwig
- Institute of Human Genetics, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Beate M Kümmerer
- Institute of Virology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, 53127, Bonn, Germany
| | - Katrin Paeschke
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- Department of Oncology, Haematology and Rheumatology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
6
|
Zhang W, Golynker I, Brosh R, Fajardo A, Zhu Y, Wudzinska AM, Ordoñez R, Ribeiro-Dos-Santos AM, Carrau L, Damani-Yokota P, Yeung ST, Khairallah C, Vela Gartner A, Chalhoub N, Huang E, Ashe HJ, Khanna KM, Maurano MT, Kim SY, tenOever BR, Boeke JD. Mouse genome rewriting and tailoring of three important disease loci. Nature 2023; 623:423-431. [PMID: 37914927 PMCID: PMC10632133 DOI: 10.1038/s41586-023-06675-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/25/2023] [Indexed: 11/03/2023]
Abstract
Genetically engineered mouse models (GEMMs) help us to understand human pathologies and develop new therapies, yet faithfully recapitulating human diseases in mice is challenging. Advances in genomics have highlighted the importance of non-coding regulatory genome sequences, which control spatiotemporal gene expression patterns and splicing in many human diseases1,2. Including regulatory extensive genomic regions, which requires large-scale genome engineering, should enhance the quality of disease modelling. Existing methods set limits on the size and efficiency of DNA delivery, hampering the routine creation of highly informative models that we call genomically rewritten and tailored GEMMs (GREAT-GEMMs). Here we describe 'mammalian switching antibiotic resistance markers progressively for integration' (mSwAP-In), a method for efficient genome rewriting in mouse embryonic stem cells. We demonstrate the use of mSwAP-In for iterative genome rewriting of up to 115 kb of a tailored Trp53 locus, as well as for humanization of mice using 116 kb and 180 kb human ACE2 loci. The ACE2 model recapitulated human ACE2 expression patterns and splicing, and notably, presented milder symptoms when challenged with SARS-CoV-2 compared with the existing K18-hACE2 model, thus representing a more human-like model of infection. Finally, we demonstrated serial genome writing by humanizing mouse Tmprss2 biallelically in the ACE2 GREAT-GEMM, highlighting the versatility of mSwAP-In in genome writing.
Collapse
Affiliation(s)
- Weimin Zhang
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Ilona Golynker
- Department of Microbiology, NYU Langone Health, New York, NY, USA
| | - Ran Brosh
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Alvaro Fajardo
- Department of Microbiology, NYU Langone Health, New York, NY, USA
| | - Yinan Zhu
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Aleksandra M Wudzinska
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Raquel Ordoñez
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - André M Ribeiro-Dos-Santos
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Lucia Carrau
- Department of Microbiology, NYU Langone Health, New York, NY, USA
| | | | - Stephen T Yeung
- Department of Microbiology, NYU Langone Health, New York, NY, USA
| | | | - Antonio Vela Gartner
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Noor Chalhoub
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Emily Huang
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Hannah J Ashe
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Kamal M Khanna
- Department of Microbiology, NYU Langone Health, New York, NY, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Matthew T Maurano
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Sang Yong Kim
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Benjamin R tenOever
- Department of Microbiology, NYU Langone Health, New York, NY, USA
- Department of Medicine, NYU Langone Health, New York, NY, USA
| | - Jef D Boeke
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA.
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA.
| |
Collapse
|
7
|
Gioukaki C, Georgiou A, Gkaralea LE, Kroupis C, Lazaris AC, Alamanis C, Thomopoulou GE. Unravelling the Role of P300 and TMPRSS2 in Prostate Cancer: A Literature Review. Int J Mol Sci 2023; 24:11299. [PMID: 37511059 PMCID: PMC10379122 DOI: 10.3390/ijms241411299] [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: 05/31/2023] [Revised: 06/26/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer is one of the most common malignant diseases in men, and it contributes significantly to the increased mortality rate in men worldwide. This study aimed to review the roles of p300 and TMPRSS2 (transmembrane protease, serine 2) in the AR (androgen receptor) pathway as they are closely related to the development and progression of prostate cancer. This paper represents a library-based study conducted by selecting the most suitable, up-to-date scientific published articles from online journals. We focused on articles that use similar techniques, particularly those that use prostate cancer cell lines and immunohistochemical staining to study the molecular impact of p300 and TMPRSS2 in prostate cancer specimens. The TMPRSS2:ERG fusion is considered relevant to prostate cancer, but its association with the development and progression as well as its clinical significance have not been fully elucidated. On the other hand, high p300 levels in prostate cancer biopsies predict larger tumor volumes, extraprostatic extension of disease, and seminal vesicle involvement at prostatectomy, and may be associated with prostate cancer progression after surgery. The inhibition of p300 has been shown to reduce the proliferation of prostate cancer cells with TMPRSS2:ETS (E26 transformation-specific) fusions, and combining p300 inhibitors with other targeted therapies may increase their efficacy. Overall, the interplay between the p300 and TMPRSS2 pathways is an active area of research.
Collapse
Affiliation(s)
- Charitomeni Gioukaki
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Alexandros Georgiou
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Christos Kroupis
- Department of Clinical Biochemistry, Attikon University Hospital, National and Kapodistrian University of Athens, 12461 Athens, Greece
| | - Andreas C Lazaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Alamanis
- 1st Urology Department, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgia Eleni Thomopoulou
- Cytopathology Department, Attikon University Hospital, National and Kapodistrian University of Athens, 12461 Athens, Greece
| |
Collapse
|
8
|
Spinello A, D'Anna L, Bignon E, Miclot T, Grandemange S, Terenzi A, Barone G, Barbault F, Monari A. Mechanism of the Covalent Inhibition of Human Transmembrane Protease Serine 2 as an Original Antiviral Strategy. J Phys Chem B 2023. [PMID: 37428676 DOI: 10.1021/acs.jpcb.3c02910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
The Transmembrane Protease Serine 2 (TMPRSS2) is a human enzyme which is involved in the maturation and post-translation of different proteins. In addition to being overexpressed in cancer cells, TMPRSS2 plays a further fundamental role in favoring viral infections by allowing the fusion of the virus envelope with the cellular membrane, notably in SARS-CoV-2. In this contribution, we resort to multiscale molecular modeling to unravel the structural and dynamical features of TMPRSS2 and its interaction with a model lipid bilayer. Furthermore, we shed light on the mechanism of action of a potential inhibitor (nafamostat), determining the free-energy profile associated with the inhibition reaction and showing the facile poisoning of the enzyme. Our study, while providing the first atomistically resolved mechanism of TMPRSS2 inhibition, is also fundamental in furnishing a solid framework for further rational design targeting transmembrane proteases in a host-directed antiviral strategy.
Collapse
Affiliation(s)
- Angelo Spinello
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
| | - Luisa D'Anna
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
| | - Emmanuelle Bignon
- Université de Lorraine and CNRS, UMR 7019 LPCT, F-54000 Nancy, France
| | - Tom Miclot
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
- Université de Lorraine and CNRS, UMR 7019 LPCT, F-54000 Nancy, France
| | | | - Alessio Terenzi
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
| | - Giampaolo Barone
- Department of Biological, Chemical and Pharmaceutical Sciences, University of Palermo, 90126 Palermo, Italy
| | | | - Antonio Monari
- Université Paris Cité and CNRS, ITODYS, F-75006 Paris, France
| |
Collapse
|
9
|
Vazquez-Rodriguez S, Ramírez-Contreras D, Noriega L, García-García A, Sánchez-Gaytán BL, Melendez FJ, Castro ME, de Azevedo WF, González-Vergara E. Interaction of copper potential metallodrugs with TMPRSS2: A comparative study of docking tools and its implications on COVID-19. Front Chem 2023; 11:1128859. [PMID: 36778030 PMCID: PMC9909424 DOI: 10.3389/fchem.2023.1128859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
SARS-CoV-2 is the virus responsible for the COVID-19 pandemic. For the virus to enter the host cell, its spike (S) protein binds to the ACE2 receptor, and the transmembrane protease serine 2 (TMPRSS2) cleaves the binding for the fusion. As part of the research on COVID-19 treatments, several Casiopeina-analogs presented here were looked at as TMPRSS2 inhibitors. Using the DFT and conceptual-DFT methods, it was found that the global reactivity indices of the optimized molecular structures of the inhibitors could be used to predict their pharmacological activity. In addition, molecular docking programs (AutoDock4, Molegro Virtual Docker, and GOLD) were used to find the best potential inhibitors by looking at how they interact with key amino acid residues (His296, Asp 345, and Ser441) in the catalytic triad. The results show that in many cases, at least one of the amino acids in the triad is involved in the interaction. In the best cases, Asp435 interacts with the terminal nitrogen atoms of the side chains in a similar way to inhibitors such as nafamostat, camostat, and gabexate. Since the copper compounds localize just above the catalytic triad, they could stop substrates from getting into it. The binding energies are in the range of other synthetic drugs already on the market. Because serine protease could be an excellent target to stop the virus from getting inside the cell, the analyzed complexes are an excellent place to start looking for new drugs to treat COVID-19.
Collapse
Affiliation(s)
- Sergio Vazquez-Rodriguez
- Centro de Química del Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Diego Ramírez-Contreras
- Centro de Química del Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Lisset Noriega
- Laboratorio de Química Teórica, Depto. de Fisicoquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico,Departamento de Física Aplicada, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mérida, Mexico
| | - Amalia García-García
- Centro de Química del Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico,Departamento de Química Inorgánica, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Brenda L. Sánchez-Gaytán
- Centro de Química del Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Francisco J. Melendez
- Laboratorio de Química Teórica, Depto. de Fisicoquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - María Eugenia Castro
- Centro de Química del Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico,*Correspondence: María Eugenia Castro, ; Enrique González-Vergara,
| | - Walter Filgueira de Azevedo
- Escola de Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Enrique González-Vergara
- Centro de Química del Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico,*Correspondence: María Eugenia Castro, ; Enrique González-Vergara,
| |
Collapse
|
10
|
Karimian A, Behjati M, Karimian M. Molecular mechanisms involved in anosmia induced by SARS-CoV-2, with a focus on the transmembrane serine protease TMPRSS2. Arch Virol 2022; 167:1931-1946. [PMID: 35939103 PMCID: PMC9358639 DOI: 10.1007/s00705-022-05545-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/31/2022] [Indexed: 11/26/2022]
Abstract
Since 2020, SARS-CoV-2 has caused a pandemic virus that has posed many challenges worldwide. Infection with this virus can result in a number of symptoms, one of which is anosmia. Olfactory dysfunction can be a temporary or long-term viral complication caused by a disorder of the olfactory neuroepithelium. Processes such as inflammation, apoptosis, and neuronal damage are involved in the development of SARS-CoV-2-induced anosmia. One of the receptors that play a key role in the entry of SARS-CoV-2 into the host cell is the transmembrane serine protease TMPRSS2, which facilitates this process by cleaving the viral S protein. The gene encoding TMPRSS2 is located on chromosome 21. It contains 15 exons and has many genetic variations, some of which increase the risk of disease. Delta strains have been shown to be more dependent on TMPRSS2 for cell entry than Omicron strains. Blockade of this receptor by serine protease inhibitors such as camostat and nafamostat can be helpful for treating SARS-CoV-2 symptoms, including anosmia. Proper understanding of the different functional aspects of this serine protease can help to overcome the therapeutic challenges of SARS-CoV-2 symptoms, including anosmia. In this review, we describe the cellular and molecular events involved in anosmia induced by SARS-CoV-2 with a focus on the function of the TMPRSS2 receptor.
Collapse
Affiliation(s)
- Ali Karimian
- Department of Otorhinolaryngology, School of Medicine, Kashan University of Medical Science, Kashan, Iran
| | - Mohaddeseh Behjati
- Cellular, Molecular and Genetics Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran.
| |
Collapse
|
11
|
Toba S, Sato A, Kawai M, Taoda Y, Unoh Y, Kusakabe S, Nobori H, Uehara S, Uemura K, Taniguchi K, Kobayashi M, Noshi T, Yoshida R, Naito A, Shishido T, Maruyama J, Paessler S, Carr MJ, Hall WW, Yoshimatsu K, Arikawa J, Matsuno K, Sakoda Y, Sasaki M, Orba Y, Sawa H, Kida H. Identification of cap-dependent endonuclease inhibitors with broad-spectrum activity against bunyaviruses. Proc Natl Acad Sci U S A 2022; 119:e2206104119. [PMID: 36037386 PMCID: PMC9457168 DOI: 10.1073/pnas.2206104119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/07/2022] [Indexed: 11/18/2022] Open
Abstract
Viral hemorrhagic fevers caused by members of the order Bunyavirales comprise endemic and emerging human infections that are significant public health concerns. Despite the disease severity, there are few therapeutic options available, and therefore effective antiviral drugs are urgently needed to reduce disease burdens. Bunyaviruses, like influenza viruses (IFVs), possess a cap-dependent endonuclease (CEN) that mediates the critical cap-snatching step of viral RNA transcription. We screened compounds from our CEN inhibitor (CENi) library and identified specific structural compounds that are 100 to 1,000 times more active in vitro than ribavirin against bunyaviruses, including Lassa virus, lymphocytic choriomeningitis virus (LCMV), and Junin virus. To investigate their inhibitory mechanism of action, drug-resistant viruses were selected in culture. Whole-genome sequencing revealed that amino acid substitutions in the CEN region of drug-resistant viruses were located in similar positions as those of the CEN α3-helix loop of IFVs derived under drug selection. Thus, our studies suggest that CENi compounds inhibit both bunyavirus and IFV replication in a mechanistically similar manner. Structural analysis revealed that the side chain of the carboxyl group at the seventh position of the main structure of the compound was essential for the high antiviral activity against bunyaviruses. In LCMV-infected mice, the compounds significantly decreased blood viral load, suppressed symptoms such as thrombocytopenia and hepatic dysfunction, and improved survival rates. These data suggest a potential broad-spectrum clinical utility of CENis for the treatment of both severe influenza and hemorrhagic diseases caused by bunyaviruses.
Collapse
Affiliation(s)
- Shinsuke Toba
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Akihiko Sato
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | | | | | - Yuto Unoh
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Shinji Kusakabe
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | | | | | - Kentaro Uemura
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Keiichi Taniguchi
- Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | | | | | | | | | | | - Junki Maruyama
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Michael J. Carr
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- National Virus Reference Laboratory, School of Medicine, University College Dublin, Dublin, Ireland
| | - William W. Hall
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- National Virus Reference Laboratory, School of Medicine, University College Dublin, Dublin, Ireland
- Global Virus Network, Baltimore, MD 21201
| | - Kumiko Yoshimatsu
- Laboratory of Animal Experimentation, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido 060-0815, Japan
| | - Jiro Arikawa
- Office for Biosafety Auditor, Nagasaki University, Nagasaki 852-8521, Japan
| | - Keita Matsuno
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Yoshihiro Sakoda
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Global Virus Network, Baltimore, MD 21201
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Hiroshi Kida
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| |
Collapse
|
12
|
Wettstein L, Kirchhoff F, Münch J. The Transmembrane Protease TMPRSS2 as a Therapeutic Target for COVID-19 Treatment. Int J Mol Sci 2022; 23:1351. [PMID: 35163273 PMCID: PMC8836196 DOI: 10.3390/ijms23031351] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 01/25/2023] Open
Abstract
TMPRSS2 is a type II transmembrane protease with broad expression in epithelial cells of the respiratory and gastrointestinal tract, the prostate, and other organs. Although the physiological role of TMPRSS2 remains largely elusive, several endogenous substrates have been identified. TMPRSS2 serves as a major cofactor in SARS-CoV-2 entry, and primes glycoproteins of other respiratory viruses as well. Consequently, inhibiting TMPRSS2 activity is a promising strategy to block viral infection. In this review, we provide an overview of the role of TMPRSS2 in the entry processes of different respiratory viruses. We then review the different classes of TMPRSS2 inhibitors and their clinical development, with a focus on COVID-19 treatment.
Collapse
Affiliation(s)
| | | | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (L.W.); (F.K.)
| |
Collapse
|
13
|
Identification of lead inhibitors of TMPRSS2 isoform 1 of SARS-CoV-2 target using neural network, random forest, and molecular docking. DATA SCIENCE FOR COVID-19 2022. [PMCID: PMC8988906 DOI: 10.1016/b978-0-323-90769-9.00021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was responsible for over 4 million confirmed cases of severe acute respiratory syndrome, of which more than 300,000 cases were confirmed to be dead as of May 2020. The virulent endocytotic activities of SARS-CoV-2 have been associated with angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2). Previous studies on the viral activation of TMPRSS2 focused most often than not on the isoform 2 of TMPRSS2, but the isoform 1 (529 residues) has also been shown to be expressed in target cells and contribute to viral activation in host. The inhibition of TMPRSS2 has been reported to grossly reduce the pathogenic effects of SARS-CoV-2 endocytotic activities. In this study therefore, we developed two machine learning models using random forest classifier (RFC) and neural networks (NNs) based on 2251 serine protease inhibitors to screen a database of 21,000,000 virtual compounds. We screened the hit compounds using absorption, distribution, metabolism, and excretion (ADME) properties and finally docked the filtered compounds into the predicted binding site of TMPRSS2 isoform 1 homology model to determine their corresponding binding affinity and plausible molecular interactions. One (ASONN) and four (ASOIRFC1–4) lead compounds were obtained from the ADME-NN and RFC filtered hits, respectively, having better binding affinity and lead-likeness properties than those of camostat; this could be due to extensive hydrogen and hydrophobic interactions.
Collapse
|
14
|
Jafarpour R, Pashangzadeh S, Dowran R. Host factors: Implications in immunopathogenesis of COVID-19. Pathol Res Pract 2021; 228:153647. [PMID: 34749207 PMCID: PMC8505027 DOI: 10.1016/j.prp.2021.153647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a viral disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is more serious in people with underlying diseases, but the cause of healthy people with progressive disease is largely unknown. Host genetic factors such as ACE2 variants, IFITM-3, HLA, TMRSS2, and furin polymorphisms appear to be one of the agents involved in the progression of the COVID-19 and outcome of the disease. This review discusses the general characteristics of SARS-CoV-2, including viral features, receptors, cell entry, clinical findings, and the main human genetic factors that may contribute to the pathogenesis of COVID-19 and get the patients' situation more complex. Further knowledge in this context may help to find a way to prevent and treat this viral pneumonia.
Collapse
Affiliation(s)
- Roghayeh Jafarpour
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Salar Pashangzadeh
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran,Immunology Today, Universal Scientific Education and Research Network (USERN), Tehan, Iran
| | - Razieh Dowran
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran,Corresponding author at: Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Wu J, Zhang L, Wang X. Host Sex Steroids Interact With Virus Infection: New Insights Into Sex Disparity in Infectious Diseases. Front Microbiol 2021; 12:747347. [PMID: 34803967 PMCID: PMC8600311 DOI: 10.3389/fmicb.2021.747347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/27/2021] [Indexed: 01/08/2023] Open
Abstract
Sex hormones are steroid hormones synthesized from the gonads of animals and tissues such as the placenta and adrenocortical reticular zone. The physiological functions of sex hormones are complex. Sex hormones are not only pathologically correlated with many diseases of the reproductive system, but are etiological factors in some viral infectious diseases, including disease caused by infections of coronaviruses, herpesviruses, hepatitis viruses, and other kinds of human viruses, which either exhibit a male propensity in clinical practice, or crosstalk with androgen receptor (AR)-related pathways in viral pathogenesis. Due to the global pandemic of coronavirus disease 2019 (COVID-19), the role of androgen/AR in viral infectious disease is highlighted again, majorly representing by the recent advances of AR-responsive gene of transmembrane protease/serine subfamily member 2 (TMPRSS2), which proteolytically activates the receptor-mediated virus entry by many coronaviruses and influenza virus, along with the role of androgen-mediated signaling for the transcription of hepatitis B virus (HBV), and the role of sex hormone responsive genes during Zika virus (ZIKV) pathogenesis, et al. Collectively, we propose to provide a comprehensive overview of the role of male sex hormones during multiple phases in the life cycle of different human viruses, which may be partly responsible for the sex-specific prevalence, severity and mortality of some diseases, therefore, may provide clues to develop more efficient prevention and treatment strategies for high-risk populations.
Collapse
Affiliation(s)
- Jinfeng Wu
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Lei Zhang
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xing Wang
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
16
|
Chan SW, Shafi T, Ford RC. Kite-Shaped Molecules Block SARS-CoV-2 Cell Entry at a Post-Attachment Step. Viruses 2021; 13:v13112306. [PMID: 34835112 PMCID: PMC8619434 DOI: 10.3390/v13112306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Anti-viral small molecules are currently lacking for treating coronavirus infection. The long development timescales for such drugs are a major problem, but could be shortened by repurposing existing drugs. We therefore screened a small library of FDA-approved compounds for potential severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antivirals using a pseudovirus system that allows a sensitive read-out of infectivity. A group of structurally-related compounds, showing moderate inhibitory activity with IC50 values in the 2–5 μM range, were identified. Further studies demonstrated that these “kite-shaped” molecules were surprisingly specific for SARS-CoV-1 and SARS-CoV-2 and that they acted early in the entry steps of the viral infectious cycle, but did not affect virus attachment to the cells. Moreover, the compounds were able to prevent infection in both kidney- and lung-derived human cell lines. The structural homology of the hits allowed the production of a well-defined pharmacophore that was found to be highly accurate in predicting the anti-viral activity of the compounds in the screen. We discuss the prospects of repurposing these existing drugs for treating current and future coronavirus outbreaks.
Collapse
|
17
|
Overcoming Culture Restriction for SARS-CoV-2 in Human Cells Facilitates the Screening of Compounds Inhibiting Viral Replication. Antimicrob Agents Chemother 2021; 65:e0009721. [PMID: 33903110 PMCID: PMC8406809 DOI: 10.1128/aac.00097-21] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Efforts to mitigate the coronavirus disease 2019 (COVID-19) pandemic include the screening of existing antiviral molecules that could be repurposed to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Although SARS-CoV-2 replicates and propagates efficiently in African green monkey kidney (Vero) cells, antivirals such as nucleos(t)ide analogs (NUCs) often show decreased activity in these cells due to inefficient metabolization. SARS-CoV-2 exhibits low viability in human cells in culture. Here, serial passages of a SARS-CoV-2 isolate (original-SARS2) in the human hepatoma cell clone Huh7.5 led to the selection of a variant (adapted-SARS2) with significantly improved infectivity in human liver (Huh7 and Huh7.5) and lung cancer (unmodified Calu-1 and A549) cells. The adapted virus exhibited mutations in the spike protein, including a 9-amino-acid deletion and 3 amino acid changes (E484D, P812R, and Q954H). E484D also emerged in Vero E6-cultured viruses that became viable in A549 cells. Original and adapted viruses were susceptible to scavenger receptor class B type 1 (SR-B1) receptor blocking, and adapted-SARS2 exhibited significantly less dependence on ACE2. Both variants were similarly neutralized by COVID-19 convalescent-phase plasma, but adapted-SARS2 exhibited increased susceptibility to exogenous type I interferon. Remdesivir inhibited original- and adapted-SARS2 similarly, demonstrating the utility of the system for the screening of NUCs. Among the tested NUCs, only remdesivir, molnupiravir, and, to a limited extent, galidesivir showed antiviral effects across human cell lines, whereas sofosbuvir, ribavirin, and favipiravir had no apparent activity. Analogously to the emergence of spike mutations in vivo, the spike protein is under intense adaptive selection pressure in cell culture. Our results indicate that the emergence of spike mutations will most likely not affect the activity of remdesivir.
Collapse
|
18
|
Pattanashetti L, Patil S, Nyamgouda S, Bhagiratha M, Gadad P. COVID-19 and pregnant women - An overview on diagnosis, treatment approach with limitation, and clinical management. Monaldi Arch Chest Dis 2021; 91. [PMID: 34121377 DOI: 10.4081/monaldi.2021.1785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/15/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease or more popularly called COVID-19 is known to be caused by a novel coronavirus 2. The COVID-19 has been identified to be originated from Wuhan, Hubei, China. This pandemic started in December 2019, and since then it has spread across the world within a short period. The health and family welfare ministry of the Government of India reported 227,546 active, 9,997,272 discharged cases, and 150,114 deaths due to COVID-19 as of 06 January 2021. Indian Council of Medical Research (ICMR) reports that the cumulative testing status of SARS-CoV-2 (COVID-19) was 931,408 up to November 03, 2020. Currently, no specific anti-viral drug for COVID-19 management is recommended in the current scenario. Vulnerable populations such as pregnant women affected by COVID-19 infection need to be recognized and followed up for effective handling concerning morbidity and mortality. At present, very few case reports on COVID-19 infected pregnant women have been published in India and there is no proven exclusive treatment protocol. This article summarizes a review of signs and symptoms, etiopathogenesis, risk factors, diagnosis, and possible management of COVID-19 infection in pregnant women. This overview may be useful for health care providers for practical approach and limitation of drugs used in the current management and considers the choice of drugs with their special attention given to adverse effects to improvise maternal health, pregnancy, and birth outcomes.
Collapse
Affiliation(s)
- Laxmi Pattanashetti
- Department of Pharmacology, KLE College of Pharmacy, Hubli (A constituent unit of KLE Academy of Higher Education and Research, Belagavi), Karnataka.
| | - Santosh Patil
- Department of Pharmacology, KLE College of Pharmacy, Hubli (A constituent unit of KLE Academy of Higher Education and Research, Belagavi), Karnataka.
| | - Sanath Nyamgouda
- Department of Pharmacy Practice, KLE College of Pharmacy, Hubli (A constituent unit of KLE Academy of Higher Education and Research, Belagavi, Karnataka.
| | - Mahendrakumar Bhagiratha
- Department of Pharmacy Practice, KLE College of Pharmacy, Hubli (A constituent unit of KLE Academy of Higher Education and Research, Belagavi, Karnataka.
| | - Pramod Gadad
- Department of Pharmacology, KLE College of Pharmacy, Hubli (A constituent unit of KLE Academy of Higher Education and Research, Belagavi), Karnataka.
| |
Collapse
|
19
|
Bukowska-Ośko I, Popiel M, Kowalczyk P. The Immunological Role of the Placenta in SARS-CoV-2 Infection-Viral Transmission, Immune Regulation, and Lactoferrin Activity. Int J Mol Sci 2021; 22:5799. [PMID: 34071527 PMCID: PMC8198160 DOI: 10.3390/ijms22115799] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
A pandemic of acute respiratory infections, due to a new type of coronavirus, can cause Severe Acute Respiratory Syndrome 2 (SARS-CoV-2) and has created the need for a better understanding of the clinical, epidemiological, and pathological features of COVID-19, especially in high-risk groups, such as pregnant women. Viral infections in pregnant women may have a much more severe course, and result in an increase in the rate of complications, including spontaneous abortion, stillbirth, and premature birth-which may cause long-term consequences in the offspring. In this review, we focus on the mother-fetal-placenta interface and its role in the potential transmission of SARS-CoV-2, including expression of viral receptors and proteases, placental pathology, and the presence of the virus in neonatal tissues and fluids. This review summarizes the current knowledge on the anti-viral activity of lactoferrin during viral infection in pregnant women, analyzes its role in the pathogenicity of pandemic virus particles, and describes the potential evidence for placental blocking/limiting of the transmission of the virus.
Collapse
Affiliation(s)
- Iwona Bukowska-Ośko
- Department of Immunopathology of Infectious and Parasitic Diseases, Medical University of Warsaw, 02-091Warsaw, Poland;
| | - Marta Popiel
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| | - Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| |
Collapse
|
20
|
Monticelli M, Hay Mele B, Benetti E, Fallerini C, Baldassarri M, Furini S, Frullanti E, Mari F, GEN-COVID Multicenter Study, Andreotti G, Cubellis MV, Renieri A. Protective Role of a TMPRSS2 Variant on Severe COVID-19 Outcome in Young Males and Elderly Women. Genes (Basel) 2021; 12:596. [PMID: 33921689 PMCID: PMC8073081 DOI: 10.3390/genes12040596] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023] Open
Abstract
The protease encoded by the TMPRSS2 gene facilitates viral infections and has been implicated in the pathogenesis of SARS-CoV-2. We analyzed the TMPRSS2 sequence and correlated the protein variants with the clinical features of a cohort of 1177 patients affected by COVID-19 in Italy. Nine relatively common variants (allele frequency > 0.01) and six missense variants which may affect the protease activity according to PolyPhen-2 in HumVar-trained mode were identified. Among them, p.V197M (p.Val197Met) (rs12329760) emerges as a common variant that has a deleterious effect on the protease and a protective effect on the patients. Its role appears particularly relevant in two subgroups of patients-young males and elderly women-and among those affected by co-morbidities, where the variant frequency is higher among individuals who were mildly affected by the disease and did not need hospitalization or oxygen therapy than among those more severely affected, who required oxygen therapy, ventilation or intubation. This study provides useful information for the identification of patients at risk of developing a severe form of COVID-19, and encourages the usage of drugs affecting the expression of TMPRSS2 or inhibiting protein activity.
Collapse
Affiliation(s)
- Maria Monticelli
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
| | - Bruno Hay Mele
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
- Integrative Marine Ecology Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy
| | - Elisa Benetti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
| | - Chiara Fallerini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Margherita Baldassarri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Mari
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| | | | | | - Maria Vittoria Cubellis
- Department of Biology, Università Federico II, 80126 Napoli, Italy; (M.M.); (B.H.M.)
- Istituto di Chimica Biomolecolare—CNR, 80078 Pozzuoli, Italy
| | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (C.F.); (M.B.); (S.F.); (E.F.); (F.M.); (A.R.)
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy
| |
Collapse
|
21
|
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has had a massive impact on human lives worldwide. While the airborne SARS-CoV-2 primarily affects the lungs, viremia is not uncommon. As placental trophoblasts are directly bathed in maternal blood, they are vulnerable to SARS-CoV-2. Intriguingly, the human fetus is largely spared from SARS-CoV-2 infection. We tested whether the human placenta expresses the main SARS-CoV-2 entry factors angiotensin-converting enzyme 2 (ACE2), transmembrane protease serine 2 (TMPRSS2), and furin and showed that ACE2 and TMPRSS2 are expressed in the trophoblast rather than in other placental villous cells. While furin is expressed in the main placental villous cell types, we surveyed, trophoblasts exhibit the highest expression. In line with the expression of these entry factors, we demonstrated that a SARS-CoV-2 pseudovirus could enter primary human trophoblasts. Mechanisms underlying placental defense against SARS-CoV-2 infection likely involve postentry processing, which may be germane for mitigating interventions against SARS-CoV-2. IMPORTANCE Pregnant women worldwide have been affected by COVID-19. As the virus is commonly spread to various organs via the bloodstream and because human placental trophoblasts are directly bathed in maternal blood, feto-placental infection by SARS-CoV-2 seems likely. However, despite the heightened risk to pregnant women, thus far the transmission risk of COVID-19 to the feto-placental unit seems extremely low. This has been recently attributed to a negligible expression of SARS-CoV-2 entry factors in the human placenta. We therefore sought to explore the expression of the entry factors ACE2 and TMPRSS2 in the different cell types of human placental villi. Using a combination of transcriptome sequencing (RNA-seq), real-time quantitative PCR (RT-qPCR), in situ hybridization, and immunofluorescence, we found that trophoblasts, but not the other main villous cell types, express ACE2 and TMPRSS2, with a broad expression of furin. Correspondingly, we also showed that primary human trophoblasts are permissive to entry of SARS-CoV-2 pseudovirus particles.
Collapse
|
22
|
Senapati S, Banerjee P, Bhagavatula S, Kushwaha PP, Kumar S. Contributions of human ACE2 and TMPRSS2 in determining host-pathogen interaction of COVID-19. J Genet 2021; 100:12. [PMID: 33707363 PMCID: PMC7904510 DOI: 10.1007/s12041-021-01262-w] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 02/05/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is at present an emerging global public health crisis. Angiotensin converting enzyme 2 (ACE2) and trans-membrane protease serine 2 (TMPRSS2) are the two major host factors that contribute to the virulence of SARS-CoV-2 and pathogenesis of coronavirus disease-19 (COVID-19). Transmission of SARS-CoV-2 from animal to human is considered a rare event that necessarily requires strong evolutionary adaptations. Till date no other human cellular receptors are identified beside ACE2 for SARS-CoV-2 entry inside the human cell. Proteolytic cleavage of viral spike (S)-protein and ACE2 by TMPRSS2 began the entire host-pathogen interaction initiated with the physical binding of ACE2 to S-protein. SARS-CoV-2 S-protein binds to ACE2 with much higher affinity and stability than that of SARS-CoVs. Molecular interactions between ACE2-S and TMPRSS2-S are crucial and preciously mediated by specific residues. Structural stability, binding affinity and level of expression of these three interacting proteins are key susceptibility factors for COVID-19. Specific protein-protein interactions (PPI) are being identified that explains uniqueness of SARS-CoV-2 infection. Amino acid substitutions due to naturally occurring genetic polymorphisms potentially alter these PPIs and poses further clinical heterogeneity of COVID-19. Repurposing of several phytochemicals and approved drugs against ACE2, TMPRSS2 and S-protein have been proposed that could inhibit PPI between them. We have also identified some novel lead phytochemicals present in Azadirachta indica and Aloe barbadensis which could be utilized for further in vitro and in vivo anti-COVID-19 drug discovery. Uncovering details of ACE2-S and TMPRSS2-S interactions would further contribute to future research on COVID-19.
Collapse
Affiliation(s)
- Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda 151 401, India.
| | | | | | | | | |
Collapse
|
23
|
Mohammadpour S, Torshizi Esfahani A, Halaji M, Lak M, Ranjbar R. An updated review of the association of host genetic factors with susceptibility and resistance to COVID-19. J Cell Physiol 2021; 236:49-54. [PMID: 32542735 PMCID: PMC7323230 DOI: 10.1002/jcp.29868] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/28/2020] [Indexed: 12/18/2022]
Abstract
The emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in human populations sparked a global pandemic of the coronavirus disease 2019 (COVID-19). According to preliminary data, about 14% of cases are considered severe and 5% of cases result in critical illness and, reported case fatality rates vary from 1% to more than 7%. However, the symptoms of the disease and the clinical outcome are very different in infected people. In view of these differences, it is clearly apparent that to gain insight into the biology of the SARS-CoV-2, it is important to study not just the infectious particle in itself but also to investigate the virus-host cell interactions that occur during infection. This review seeks to consider the various aspects of genetic factors in determining the susceptibility and host resistance to SARS-CoV-2 throughout the recently published literature.
Collapse
Affiliation(s)
| | - Amir Torshizi Esfahani
- Department of Biology, Faculty of Biological SciencesIslamic Azad University, North Tehran BranchTehranIran
| | - Mehrdad Halaji
- Department of Microbiology, School of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Mina Lak
- Department of Microbiology, Faculty of Advanced Science and TechnologyIslamic Azad University Tehran Medical SciencesTehranIran
| | - Reza Ranjbar
- Molecular Biology Research Center, Systems Biology and Poisonings InstituteBaqiyatallah University of Medical SciencesTehranIran
| |
Collapse
|
24
|
El Amri C. Serine Protease Inhibitors to Treat Lung Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:215-226. [PMID: 34019272 DOI: 10.1007/978-3-030-68748-9_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lung is a vital organ that ensures breathing function. It provides the essential interface of air filtering providing oxygen to the whole body and eliminating carbon dioxide in the blood; because of its exposure to the external environment, it is fall prey to many exogenous elements, such as pathogens, especially viral infections or environmental toxins and chemicals. These exogenous actors in addition to intrinsic disorders lead to important inflammatory responses that compromise lung tissue and normal functioning. Serine proteases regulating inflammation responses are versatile enzymes, usually involved in pro-inflammatory cytokines or other molecular mediator's production and activation of immune cells. In this chapter, an overview on major serine proteases in airway inflammation as therapeutic targets and their clinically relevant inhibitors is provided. Recent updates on serine protease inhibitors in the context of the COVID-19 pandemic are summarized.
Collapse
Affiliation(s)
- Chahrazade El Amri
- Sorbonne Université, Faculty of Sciences and Engineering, IBPS, UMR 8256 CNRS-UPMC, ERL INSERM U1164, Biological Adaptation and Ageing, Paris, France.
| |
Collapse
|
25
|
Cocozza F, Névo N, Piovesana E, Lahaye X, Buchrieser J, Schwartz O, Manel N, Tkach M, Théry C, Martin‐Jaular L. Extracellular vesicles containing ACE2 efficiently prevent infection by SARS-CoV-2 Spike protein-containing virus. J Extracell Vesicles 2020; 10:e12050. [PMID: 33391636 PMCID: PMC7769856 DOI: 10.1002/jev2.12050] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/16/2020] [Accepted: 12/04/2020] [Indexed: 12/29/2022] Open
Abstract
SARS-CoV-2 entry is mediated by binding of the spike protein (S) to the surface receptor ACE2 and subsequent priming by host TMPRSS2 allowing membrane fusion. Here, we produced extracellular vesicles (EVs) exposing ACE2 and demonstrate that ACE2-EVs are efficient decoys for SARS-CoV-2 S protein-containing lentivirus. Reduction of infectivity positively correlates with the level of ACE2, is much more efficient than with soluble ACE2 and further enhanced by the inclusion of TMPRSS2.
Collapse
Affiliation(s)
- Federico Cocozza
- INSERM U932Institut Curie Centre de RecherchePSL Research UniversityParisFrance
- Université de ParisParisFrance
| | - Nathalie Névo
- INSERM U932Institut Curie Centre de RecherchePSL Research UniversityParisFrance
| | - Ester Piovesana
- INSERM U932Institut Curie Centre de RecherchePSL Research UniversityParisFrance
- Université de ParisParisFrance
| | - Xavier Lahaye
- INSERM U932Institut Curie Centre de RecherchePSL Research UniversityParisFrance
| | - Julian Buchrieser
- Virus and Immunity UnitInstitut Pasteur and CNRS UMR 3569ParisFrance
| | - Olivier Schwartz
- Virus and Immunity UnitInstitut Pasteur and CNRS UMR 3569ParisFrance
| | - Nicolas Manel
- INSERM U932Institut Curie Centre de RecherchePSL Research UniversityParisFrance
| | - Mercedes Tkach
- INSERM U932Institut Curie Centre de RecherchePSL Research UniversityParisFrance
| | - Clotilde Théry
- INSERM U932Institut Curie Centre de RecherchePSL Research UniversityParisFrance
| | | |
Collapse
|
26
|
Abstract
Transmembrane serine protease 2 is encoded by the TMPRSS2 gene. The gene is widely conserved and has two isoforms, both being autocatalytically activated from the inactive zymogen form. A fusion gene between the TMPRSS2 gene and ERG (erythroblast-specific-related gene), an oncogenic transcription factor, is the most common chromosomal aberration detected in prostate cancer, responsible for driving carcinogenesis. The other key role of TMPRSS2 is in priming the viral spike protein which facilitates viral entry essential for viral infectivity. The protease activates a diverse range of viruses. Both SARS-CoV and SARS-CoV-2 (COVID-19) use angiotensin-converting enzyme 2 (ACE2) and TMPRSS2 to facilitate entry to cells, but with SARS-CoV-2 human-to-human transmission is much higher than SARS-CoV. As TMPRSS2 is expressed outside of the lung, and can therefore contribute to extrapulmonary spread of viruses, it warrants further exploration as a potential target for limiting viral spread and infectivity.
Collapse
Affiliation(s)
- Michelle Thunders
- Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Brett Delahunt
- Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| |
Collapse
|
27
|
Pollard CA, Morran MP, Nestor-Kalinoski AL. The COVID-19 pandemic: a global health crisis. Physiol Genomics 2020; 52:549-557. [PMID: 32991251 PMCID: PMC7686876 DOI: 10.1152/physiolgenomics.00089.2020] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/22/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
The novel coronavirus SARS-CoV-2 was identified as the causative agent for a series of atypical respiratory diseases in the Hubei Province of Wuhan, China in December of 2019. The disease SARS-CoV-2, termed COVID-19, was officially declared a pandemic by the World Health Organization on March 11, 2020. SARS-CoV-2 contains a single-stranded, positive-sense RNA genome surrounded by an extracellular membrane containing a series of spike glycoproteins resembling a crown. COVID-19 infection results in diverse symptoms and morbidity depending on individual genetics, ethnicity, age, and geographic location. In severe cases, COVID-19 pathophysiology includes destruction of lung epithelial cells, thrombosis, hypercoagulation, and vascular leak leading to sepsis. These events lead to acute respiratory distress syndrome (ARDS) and subsequent pulmonary fibrosis in patients. COVID-19 risk factors include cardiovascular disease, hypertension, and diabetes, which are highly prevalent in the United States. This population has upregulation of the angiotensin converting enzyme-2 (ACE2) receptor, which is exploited by COVID-19 as the route of entry and infection. Viral envelope proteins bind to and degrade ACE2 receptors, thus preventing normal ACE2 function. COVID-19 infection causes imbalances in ACE2 and induces an inflammatory immune response, known as a cytokine storm, both of which amplify comorbidities within the host. Herein, we discuss the genetics, pathogenesis, and possible therapeutics of COVID-19 infection along with secondary complications associated with disease progression, including ARDS and pulmonary fibrosis. Understanding the mechanisms of COVID-19 infection will allow the development of vaccines or other novel therapeutic approaches to prevent transmission or reduce the severity of infection.
Collapse
Affiliation(s)
- Casey A Pollard
- Department of Surgery, The University of Toledo, College of Medicine and Life Sciences, Toledo, Ohio
| | - Michael P Morran
- Department of Surgery, The University of Toledo, College of Medicine and Life Sciences, Toledo, Ohio
- The University of Toledo Advanced Microscopy and Imaging Center, The University of Toledo, College of Medicine and Life Sciences, Toledo, Ohio
| | - Andrea L Nestor-Kalinoski
- Department of Surgery, The University of Toledo, College of Medicine and Life Sciences, Toledo, Ohio
- The University of Toledo Advanced Microscopy and Imaging Center, The University of Toledo, College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
28
|
Alberca RW, Pereira NZ, Oliveira LMDS, Gozzi-Silva SC, Sato MN. Pregnancy, Viral Infection, and COVID-19. Front Immunol 2020; 11:1672. [PMID: 32733490 PMCID: PMC7358375 DOI: 10.3389/fimmu.2020.01672] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Pregnancy comprises a unique immunological condition, to allow fetal development and to protect the host from pathogenic infections. Viral infections during pregnancy can disrupt immunological tolerance and may generate deleterious effects on the fetus. Despite these possible links between pregnancy and infection-induced morbidity, it is unclear how pregnancy interferes with maternal response to some viral pathogens. In this context, the novel coronavirus (SARS-CoV-2) can induce the coronavirus diseases-2019 (COVID-19) in pregnant women. The potential risk of vertical transmission is unclear, babies born from COVID-19-positive mothers seems to have no serious clinical symptoms, the possible mechanisms are discussed, which highlights that checking the children's outcome and more research is warranted. In this review, we investigate the reports concerning viral infections and COVID-19 during pregnancy, to establish a correlation and possible implications of COVID-19 during pregnancy and neonatal's health.
Collapse
MESH Headings
- Betacoronavirus
- COVID-19
- Child, Preschool
- Coronavirus Infections/blood
- Coronavirus Infections/immunology
- Coronavirus Infections/transmission
- Coronavirus Infections/virology
- Cytokines/blood
- Female
- Fetal Development/immunology
- Humans
- Infant
- Infant, Newborn
- Infectious Disease Transmission, Vertical
- Mothers
- Pandemics
- Pneumonia, Viral/blood
- Pneumonia, Viral/immunology
- Pneumonia, Viral/transmission
- Pneumonia, Viral/virology
- Pregnancy
- Pregnancy Complications, Infectious/blood
- Pregnancy Complications, Infectious/immunology
- Pregnancy Complications, Infectious/virology
- SARS-CoV-2
Collapse
Affiliation(s)
- Ricardo Wesley Alberca
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, School of Medicine and Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil
| | - Nátalli Zanete Pereira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, School of Medicine and Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil
| | - Luanda Mara Da Silva Oliveira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, School of Medicine and Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil
| | | | - Maria Notomi Sato
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
29
|
Rothan HA, Acharya A, Reid SP, Kumar M, Byrareddy SN. Molecular Aspects of COVID-19 Differential Pathogenesis. Pathogens 2020; 9:E538. [PMID: 32640525 PMCID: PMC7400297 DOI: 10.3390/pathogens9070538] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 01/08/2023] Open
Abstract
In the absence of therapeutic interventions, and a possible vaccine candidate, the spread of COVID-19 disease and associated fatalities are on the rise. The high mutation frequency in the genomic material of these viruses supports their ability to adapt to new environments, resulting in an efficient alteration in tissue tropism and host range. Therefore, the coronavirus' health threats could be relevant for the long-term. The epidemiological data indicate that age, sex, and cardio-metabolic disease have a significant impact on the spread and severity of COVID-19. In this review, we highlight recent updates on the pathogenesis of SARS-CoV-2 among men and women, including children. We also discuss the role of the cellular receptors and coreceptors used by the virus to enter host cells on differential infection among men, women, and cardio-metabolic patients.
Collapse
Affiliation(s)
- Hussin A. Rothan
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Centre, Omaha, NE 68198, USA;
| | - St Patrick Reid
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Centre, Omaha, NE 68198, USA;
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Centre, Omaha, NE 68198, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Centre, Omaha, NE 68198, USA
| |
Collapse
|
30
|
Zmora P, Hoffmann M, Kollmus H, Moldenhauer AS, Danov O, Braun A, Winkler M, Schughart K, Pöhlmann S. TMPRSS11A activates the influenza A virus hemagglutinin and the MERS coronavirus spike protein and is insensitive against blockade by HAI-1. J Biol Chem 2018; 293:13863-13873. [PMID: 29976755 PMCID: PMC6130959 DOI: 10.1074/jbc.ra118.001273] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/15/2018] [Indexed: 12/25/2022] Open
Abstract
The influenza virus hemagglutinin (HA) facilitates viral entry into target cells. Cleavage of HA by host cell proteases is essential for viral infectivity, and the responsible enzymes are potential targets for antiviral intervention. The type II transmembrane serine protease (TTSP) TMPRSS2 has been identified as an HA activator in cell culture and in the infected host. However, it is less clear whether TMPRSS2-related enzymes can also activate HA for spread in target cells. Moreover, the activity of cellular serine protease inhibitors against HA-activating TTSPs is poorly understood. Here, we show that TMPRSS11A, another member of the TTSP family, cleaves and activates the influenza A virus (FLUAV) HA and the Middle East respiratory syndrome coronavirus spike protein (MERS-S). Moreover, we demonstrate that TMPRSS11A is expressed in murine tracheal epithelium, which is a target of FLUAV infection, and in human trachea, suggesting that the protease could support FLUAV spread in patients. Finally, we show that HA activation by the TMPRSS11A-related enzymes human airway tryptase and DESC1, but not TMPRSS11A itself, is blocked by the cellular serine protease inhibitor hepatocyte growth factor activator inhibitor type-1 (HAI-1). Our results suggest that TMPRSS11A could promote FLUAV spread in target cells and that HA-activating TTSPs exhibit differential sensitivity to blockade by cellular serine protease inhibitors.
Collapse
Affiliation(s)
- Pawel Zmora
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany,
| | - Markus Hoffmann
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Heike Kollmus
- the Department of Infection Genetics, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Anna-Sophie Moldenhauer
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Olga Danov
- the Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of REBIRTH Cluster of Excellence, 30625 Hannover, Germany, and
| | - Armin Braun
- the Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of REBIRTH Cluster of Excellence, 30625 Hannover, Germany, and
| | - Michael Winkler
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Klaus Schughart
- the Department of Infection Genetics, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany.,the University of Veterinary Medicine Hannover, 30599 Hannover, Germany.,the Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Stefan Pöhlmann
- From the Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany, .,the Faculty of Biology and Psychology, University of Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
31
|
Magnen M, Gueugnon F, Guillon A, Baranek T, Thibault VC, Petit-Courty A, de Veer SJ, Harris J, Humbles AA, Si-Tahar M, Courty Y. Kallikrein-Related Peptidase 5 Contributes to H3N2 Influenza Virus Infection in Human Lungs. J Virol 2017; 91:e00421-17. [PMID: 28615200 PMCID: PMC5533929 DOI: 10.1128/jvi.00421-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/24/2017] [Indexed: 12/15/2022] Open
Abstract
Hemagglutinin (HA) of influenza virus must be activated by proteolysis before the virus can become infectious. Previous studies indicated that HA cleavage is driven by membrane-bound or extracellular serine proteases in the respiratory tract. However, there is still uncertainty as to which proteases are critical for activating HAs of seasonal influenza A viruses (IAVs) in humans. This study focuses on human KLK1 and KLK5, 2 of the 15 serine proteases known as the kallikrein-related peptidases (KLKs). We find that their mRNA expression in primary human bronchial cells is stimulated by IAV infection. Both enzymes cleaved recombinant HA from several strains of the H1 and/or H3 virus subtype in vitro, but only KLK5 promoted the infectivity of A/Puerto Rico/8/34 (H1N1) and A/Scotland/20/74 (H3N2) virions in MDCK cells. We assessed the ability of treated viruses to initiate influenza in mice. The nasal instillation of only the KLK5-treated virus resulted in weight loss and lethal outcomes. The secretion of this protease in the human lower respiratory tract is enhanced during influenza. Moreover, we show that pretreatment of airway secretions with a KLK5-selective inhibitor significantly reduced the activation of influenza A/Scotland/20/74 virions, providing further evidence of its importance. Differently, increased KLK1 secretion appeared to be associated with the recruitment of inflammatory cells in human airways regardless of the origin of inflammation. Thus, our findings point to the involvement of KLK5 in the proteolytic activation and spread of seasonal influenza viruses in humans.IMPORTANCE Influenza A viruses (IAVs) cause acute infection of the respiratory tract that affects millions of people during seasonal outbreaks every year. Cleavage of the hemagglutinin precursor by host proteases is a critical step in the life cycle of these viruses. Consequently, host proteases that activate HA can be considered promising targets for the development of new antivirals. However, the specific proteases that activate seasonal influenza viruses, especially H3N2 viruses, in the human respiratory tract have remain undefined despite many years of work. Here we demonstrate that the secreted, extracellular protease KLK5 (kallikrein-related peptidase 5) is efficient in promoting the infectivity of H3N2 IAV in vitro and in vivo Furthermore, we found that its secretion was selectively enhanced in the human lower respiratory tract during a seasonal outbreak dominated by an H3N2 virus. Collectively, our data support the clinical relevance of this protease in human influenza pathogenesis.
Collapse
Affiliation(s)
- Mélia Magnen
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
- Université François Rabelais, Tours, France
| | - Fabien Gueugnon
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
- Université François Rabelais, Tours, France
| | - Antoine Guillon
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
- Université François Rabelais, Tours, France
| | - Thomas Baranek
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
- Université François Rabelais, Tours, France
| | - Virginie C Thibault
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
- Université François Rabelais, Tours, France
| | - Agnès Petit-Courty
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
- Université François Rabelais, Tours, France
| | | | | | | | - Mustapha Si-Tahar
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
- Université François Rabelais, Tours, France
| | - Yves Courty
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
- Université François Rabelais, Tours, France
| |
Collapse
|
32
|
Non-human primate orthologues of TMPRSS2 cleave and activate the influenza virus hemagglutinin. PLoS One 2017; 12:e0176597. [PMID: 28493964 PMCID: PMC5426610 DOI: 10.1371/journal.pone.0176597] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/13/2017] [Indexed: 01/09/2023] Open
Abstract
The cellular serine protease TMPRSS2, a member of the type II transmembrane serine protease (TTSP) family, cleaves and activates the hemagglutinin of influenza A viruses (FLUAV) in cell culture and is essential for spread of diverse FLUAV in mice. Non-human primates (NHP), in particular rhesus and cynomolgus macaques, serve as animal models for influenza and experimental FLUAV infection of common marmosets has recently also been reported. However, it is currently unknown whether the NHP orthologues of human TMPRSS2 cleave and activate FLUAV hemagglutinin and contribute to viral spread in respiratory tissue. Here, we cloned and functionally analyzed the macaque and marmoset orthologues of human TMPRSS2. In addition, we analyzed the macaque orthologues of human TMPRSS4 and HAT, which also belong to the TTSP family. We found that all NHP orthologues of human TMPRSS2, TMPRSS4 and HAT cleave and activate HA upon directed expression and provide evidence that endogenous TMPRSS2 is expressed in the respiratory epithelium of rhesus macaques. Finally, we demonstrate that a serine protease inhibitor active against TMPRSS2 suppresses FLUAV spread in precision-cut lung slices of human, macaque and marmoset origin. These results indicate that FLUAV depends on serine protease activity for spread in diverse NHP and in humans. Moreover, our findings suggest that macaques and marmosets may serve as models to study FLUAV activation by TMPRSS2 in human patients.
Collapse
|