201
|
Zhou YW, Xie Y, Tang LS, Pu D, Zhu YJ, Liu JY, Ma XL. Therapeutic targets and interventional strategies in COVID-19: mechanisms and clinical studies. Signal Transduct Target Ther 2021; 6:317. [PMID: 34446699 PMCID: PMC8390046 DOI: 10.1038/s41392-021-00733-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/27/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Owing to the limitations of the present efforts on drug discovery against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the lack of the understanding of the biological regulation mechanisms underlying COVID-19, alternative or novel therapeutic targets for COVID-19 treatment are still urgently required. SARS-CoV-2 infection and immunity dysfunction are the two main courses driving the pathogenesis of COVID-19. Both the virus and host factors are potential targets for antiviral therapy. Hence, in this study, the current therapeutic strategies of COVID-19 have been classified into "target virus" and "target host" categories. Repurposing drugs, emerging approaches, and promising potential targets are the implementations of the above two strategies. First, a comprehensive review of the highly acclaimed old drugs was performed according to evidence-based medicine to provide recommendations for clinicians. Additionally, their unavailability in the fight against COVID-19 was analyzed. Next, a profound analysis of the emerging approaches was conducted, particularly all licensed vaccines and monoclonal antibodies (mAbs) enrolled in clinical trials against primary SARS-CoV-2 and mutant strains. Furthermore, the pros and cons of the present licensed vaccines were compared from different perspectives. Finally, the most promising potential targets were reviewed, and the update of the progress of treatments has been summarized based on these reviews.
Collapse
Affiliation(s)
- Yu-Wen Zhou
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yao Xie
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Dermatovenerology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Lian-Sha Tang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dan Pu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ya-Juan Zhu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ji-Yan Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Xue-Lei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
202
|
Lodhi N, Singh R, Rajput SP, Saquib Q. SARS-CoV-2: Understanding the Transcriptional Regulation of ACE2 and TMPRSS2 and the Role of Single Nucleotide Polymorphism (SNP) at Codon 72 of p53 in the Innate Immune Response against Virus Infection. Int J Mol Sci 2021; 22:8660. [PMID: 34445373 PMCID: PMC8395432 DOI: 10.3390/ijms22168660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/15/2022] Open
Abstract
Human ACE2 and the serine protease TMPRSS2 of novel SARS-CoV-2 are primary entry receptors in host cells. Expression of these genes at the transcriptional level has not been much discussed in detail. The ISRE elements of the ACE2 promoter are a binding site for the ISGF3 complex of the JAK/STAT signaling pathway. TMPRSS2, including IFNβ, STAT1, and STAT2, has the PARP1 binding site near to TSS either up or downstream promoter region. It is well documented that PARP1 regulates gene expression at the transcription level. Therefore, to curb virus infection, both promoting type I IFN signaling to boost innate immunity and prevention of virus entry by inhibiting PARP1, ACE2 or TMPRSS2 are safe options. Most importantly, our aim is to attract the attention of the global scientific community towards the codon 72 Single Nucleotide Polymorphism (SNP) of p53 and its underneath role in the innate immune response against SARS-CoV-2. Here, we discuss codon 72 SNP of human p53's role in the different innate immune response to restrict virus-mediated mortality rate only in specific parts of the world. In addition, we discuss potential targets and emerging therapies using bioengineered bacteriophage, anti-sense, or CRISPR strategies.
Collapse
Affiliation(s)
- Niraj Lodhi
- Clinical Research (Research and Development Division) miRNA Analytics LLC, Harlem Bio-Space, New York, NY 10027, USA
| | - Rubi Singh
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA;
| | | | - Quaiser Saquib
- Department of Zoology, College of Sciences, King Saud University, Riyadh 12372, Saudi Arabia;
| |
Collapse
|
203
|
Narożna M, Rubiś B. Anti-SARS-CoV-2 Strategies and the Potential Role of miRNA in the Assessment of COVID-19 Morbidity, Recurrence, and Therapy. Int J Mol Sci 2021; 22:8663. [PMID: 34445368 PMCID: PMC8395427 DOI: 10.3390/ijms22168663] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/07/2021] [Accepted: 08/08/2021] [Indexed: 02/06/2023] Open
Abstract
Recently, we have experienced a serious pandemic. Despite significant technological advances in molecular technologies, it is very challenging to slow down the infection spread. It appeared that due to globalization, SARS-CoV-2 spread easily and adapted to new environments or geographical or weather zones. Additionally, new variants are emerging that show different infection potential and clinical outcomes. On the other hand, we have some experience with other pandemics and some solutions in virus elimination that could be adapted. This is of high importance since, as the latest reports demonstrate, vaccine technology might not follow the new, mutated virus outbreaks. Thus, identification of novel strategies and markers or diagnostic methods is highly necessary. For this reason, we present some of the latest views on SARS-CoV-2/COVID-19 therapeutic strategies and raise a solution based on miRNA. We believe that in the face of the rapidly increasing global situation and based on analogical studies of other viruses, the possibility of using the biological potential of miRNA technology is very promising. It could be used as a promising diagnostic and prognostic factor, as well as a therapeutic target and tool.
Collapse
Affiliation(s)
- Maria Narożna
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, 4 Święcickiego St., 60-781 Poznan, Poland;
| | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznan, Poland
| |
Collapse
|
204
|
Bhavaniramya S, Ramar V, Vishnupriya S, Palaniappan R, Sibiya A, Baskaralingam V. Comprehensive analysis of SARS-COV-2 drug targets and pharmacological aspects in treating the COVID-19. Curr Mol Pharmacol 2021; 15:393-417. [PMID: 34382513 DOI: 10.2174/1874467214666210811120635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/27/2021] [Accepted: 02/22/2021] [Indexed: 11/22/2022]
Abstract
Corona viruses are enveloped, single-stranded RNA (Ribonucleic acid) viruses and they cause pandemic diseases having a devastating effect on both human healthcare and the global economy. To date, six corona viruses have been identified as pathogenic organisms which are significantly responsible for the infection and also cause severe respiratory diseases. Among them, the novel SARS-CoV-2 (Severe acute respiratory syndrome coronavirus 2) caused a major outbreak of corona virus diseases 2019 (COVID-19). Coronaviridae family members can affects both humans and animals. In human, corona viruses cause severe acute respiratory syndrome with mild to severe outcomes. Several structural and genomics have been investigated, and the genome encodes about 28 proteins most of them with unknown function though it shares remarkable sequence identity with other proteins. There is no potent and licensed vaccine against SARS-CoV-2 and several trials are underway to investigate the possible therapeutic agents against viral infection. However, some of the antiviral drugs that have been investigated against SARS-CoV-2 are under clinical trials. In the current review we comparatively emphasize the emergence and pathogenicity of the SARS-CoV-2 and their infection and discuss the various putative drug targets of both viral and host receptors for developing effective vaccines and therapeutic combinations to overcome the viral outbreak.
Collapse
Affiliation(s)
- Sundaresan Bhavaniramya
- Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Karaikudi 630004, Tamil Nadu. India
| | - Vanajothi Ramar
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620024. India
| | - Selvaraju Vishnupriya
- College of Food and Dairy Technology, Tamil Nadu Veterinary and Animal Sciences University, Chennai 600052. India
| | - Ramasamy Palaniappan
- Research and Development Wing, Sree Balaji Medical College and Hospital, Bharath Institute of Higher Education (BIHER), Chennai-600044, Tamilnadu. India
| | - Ashokkumar Sibiya
- Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Karaikudi 630004, Tamil Nadu. India
| | - Vaseeharan Baskaralingam
- Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Karaikudi 630004, Tamil Nadu. India
| |
Collapse
|
205
|
Azevedo PRG, Freitas NLDE, Brandão F. Testosterone and COVID-19 - a stone in the way. AN ACAD BRAS CIENC 2021; 93:e20210510. [PMID: 34378642 DOI: 10.1590/0001-3765202120200510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/19/2021] [Indexed: 11/21/2022] Open
Affiliation(s)
- Pedro Ricardo G Azevedo
- University of Brasília, Faculty of Health, Department of Pharmacy, Laboratory of Clinical Analysis, Campus Darcy Ribeiro, Asa Norte, 70910-900 Brasília, DF, Brazil
| | - Natália L DE Freitas
- University of Brasília, Faculty of Health, Department of Pharmacy, Laboratory of Clinical Analysis, Campus Darcy Ribeiro, Asa Norte, 70910-900 Brasília, DF, Brazil
| | - Fabiana Brandão
- University of Brasília, Faculty of Health, Department of Pharmacy, Laboratory of Clinical Analysis, Campus Darcy Ribeiro, Asa Norte, 70910-900 Brasília, DF, Brazil
| |
Collapse
|
206
|
ACE2 protein expression within isogenic cell lines is heterogeneous and associated with distinct transcriptomes. Sci Rep 2021; 11:15900. [PMID: 34354120 PMCID: PMC8342525 DOI: 10.1038/s41598-021-95308-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
The membrane protein angiotensin-converting enzyme 2 (ACE2) is a physiologic regulator of the renin-angiotensin system and the cellular receptor for the SARS-CoV-2 virus. Prior studies of ACE2 expression have primarily focused on mRNA abundance, with investigation at the protein level limited by uncertain specificity of commercial ACE2 antibodies. Here, we report our development of a sensitive and specific flow cytometry-based assay for cellular ACE2 protein abundance. Application of this approach to multiple cell lines revealed an unexpected degree of cellular heterogeneity, with detectable ACE2 protein in only a subset of cells in each isogenic population. This heterogeneity was mediated at the mRNA level by transcripts predominantly initiated from the ACE2 proximal promoter. ACE2 expression was heritable but not fixed over multiple generations of daughter cells, with gradual drift toward the original heterogeneous background. RNA-seq profiling identified distinct transcriptomes of ACE2-expressing relative cells to non-expressing cells, with enrichment in functionally related genes and transcription factor target sets. Our findings provide a validated approach for the specific detection of ACE2 protein at the surface of single cells, support an epigenetic mechanism of ACE2 gene regulation, and identify specific pathways associated with ACE2 expression in HuH7 cells.
Collapse
|
207
|
Piticchio T, Le Moli R, Tumino D, Frasca F. Relationship between betacoronaviruses and the endocrine system: a new key to understand the COVID-19 pandemic-A comprehensive review. J Endocrinol Invest 2021; 44:1553-1570. [PMID: 33583003 PMCID: PMC7882054 DOI: 10.1007/s40618-020-01486-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND A new harmful respiratory disease, called COVID-19 emerged in China in December 2019 due to the infection of a novel coronavirus, called SARS-Coronavirus 2 (SARS-CoV-2), which belongs to the betacoronavirus genus, including SARS-CoV-1 and MERS-CoV. SARS-CoV-2 shares almost 80% of the genome with SARS-CoV-1 and 50% with MERS-CoV. Moreover, SARS-CoV-2 proteins share a high degree of homology (approximately 95%) with SARS-CoV-1 proteins. Hence, the mechanisms of SARS-Cov-1 and SARS-Cov-2 infection are similar and occur via binding to ACE2 protein, which is widely distributed in the human body, with a predominant expression in endocrine tissues including testis, thyroid, adrenal and pituitary. PURPOSE On the basis of expression pattern of the ACE2 protein among different tissues, similarity between SARS-Cov-1 and SARS-Cov-2 and the pathophysiology of COVID-19 disease, we aimed at discussing, after almost one-year pandemic, about the relationships between COVID-19 infection and the endocrine system. First, we discussed the potential effect of hormones on the susceptibility to COVID-19 infection; second, we examined the evidences regarding the effect of COVID-19 on the endocrine system. When data were available, a comparative discussion between SARS and COVID-19 effects was also performed. METHODS A comprehensive literature search within Pubmed was performed. This review has been conducted according to the PRISMA statements. RESULTS Among 450, 100 articles were selected. Tissue and vascular damages have been shown on thyroid, adrenal, testis and pituitary glands, with multiple alterations of endocrine function. CONCLUSION Hormones may affect patient susceptibility to COVID-19 infection but evidences regarding therapeutic implication of these findings are still missing. SARS and COVID-19 may affect endocrine glands and their dense vascularization, impairing endocrine system function. A possible damage of endocrine system in COVID-19 patients should be investigated in both COVID-19 acute phase and recovery to identify both early and late endocrine complications that may be important for patient's prognosis and well-being after COVID-19 infection.
Collapse
Affiliation(s)
- T Piticchio
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - R Le Moli
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - D Tumino
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - F Frasca
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy.
| |
Collapse
|
208
|
Chen F, Shi Q, Pei F, Vogt A, Porritt RA, Garcia G, Gomez AC, Cheng MH, Schurdak ME, Liu B, Chan SY, Arumugaswami V, Stern AM, Taylor DL, Arditi M, Bahar I. A systems-level study reveals host-targeted repurposable drugs against SARS-CoV-2 infection. Mol Syst Biol 2021; 17:e10239. [PMID: 34339582 PMCID: PMC8328275 DOI: 10.15252/msb.202110239] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/22/2022] Open
Abstract
Understanding the mechanism of SARS-CoV-2 infection and identifying potential therapeutics are global imperatives. Using a quantitative systems pharmacology approach, we identified a set of repurposable and investigational drugs as potential therapeutics against COVID-19. These were deduced from the gene expression signature of SARS-CoV-2-infected A549 cells screened against Connectivity Map and prioritized by network proximity analysis with respect to disease modules in the viral-host interactome. We also identified immuno-modulating compounds aiming at suppressing hyperinflammatory responses in severe COVID-19 patients, based on the transcriptome of ACE2-overexpressing A549 cells. Experiments with Vero-E6 cells infected by SARS-CoV-2, as well as independent syncytia formation assays for probing ACE2/SARS-CoV-2 spike protein-mediated cell fusion using HEK293T and Calu-3 cells, showed that several predicted compounds had inhibitory activities. Among them, salmeterol, rottlerin, and mTOR inhibitors exhibited antiviral activities in Vero-E6 cells; imipramine, linsitinib, hexylresorcinol, ezetimibe, and brompheniramine impaired viral entry. These novel findings provide new paths for broadening the repertoire of compounds pursued as therapeutics against COVID-19.
Collapse
Affiliation(s)
- Fangyuan Chen
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- School of MedicineTsinghua UniversityBeijingChina
| | - Qingya Shi
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- School of MedicineTsinghua UniversityBeijingChina
| | - Fen Pei
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Andreas Vogt
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Rebecca A Porritt
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
- Biomedical Sciences, Infectious and Immunologic Diseases Research CenterCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Gustavo Garcia
- Department of Molecular and Medical PharmacologyDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of CaliforniaLos AngelesCAUSA
| | - Angela C Gomez
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Mary Hongying Cheng
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Mark E Schurdak
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Bing Liu
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Stephen Y Chan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine InstituteUniversity of Pittsburgh Medical CenterPittsburghPAUSA
- Division of CardiologyDepartment of MedicineUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical PharmacologyDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of CaliforniaLos AngelesCAUSA
| | - Andrew M Stern
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - D Lansing Taylor
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| | - Moshe Arditi
- Department of PediatricsDivision of Pediatric Infectious Diseases and ImmunologyCedars‐Sinai Medical CenterLos AngelesCAUSA
- Biomedical Sciences, Infectious and Immunologic Diseases Research CenterCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Ivet Bahar
- Department of Computational and Systems BiologySchool of MedicineUniversity of PittsburghPittsburghPAUSA
- University of Pittsburgh Drug Discovery InstitutePittsburghPAUSA
| |
Collapse
|
209
|
Murugan NA, Pandian CJ, Jeyakanthan J. Computational investigation on Andrographis paniculata phytochemicals to evaluate their potency against SARS-CoV-2 in comparison to known antiviral compounds in drug trials. J Biomol Struct Dyn 2021; 39:4415-4426. [PMID: 32543978 PMCID: PMC7309306 DOI: 10.1080/07391102.2020.1777901] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022]
Abstract
The outbreak due to SARS-CoV-2 (or Covid-19) is spreading alarmingly and number of deaths due to infection is aggressively increasing every day. Due to the rapid human to human transmission of Covid-19, we are in need to find a potent drug at the earliest by ruling-out the traditional time-consuming approach of drug development. This is only possible if we use reliable computational approaches for screening compounds from chemical space or by drug repurposing or by finding the phytochemicals and nutraceuticals from plants as they can be immediately used without the need for carrying out drug-trials to test safety and efficacy. A number of plant products were routinely suggested as drugs in traditional Indian and Chinese medicine. Here using molecular docking approach, and combined molecular dynamics and MM-GBSA based free energy calculations approach, we study the potency of the four selected phytochemicals namely andrographolide (AGP1), 14-deoxy 11,12-didehydro andrographolide (AGP2), neoandrographolide (AGP3) and 14-deoxy andrographolide (AGP4) from A. paniculata plant against the four key targets including three non-structural proteins (3 L main protease (3CLpro), Papain-like proteinase (PLpro) and RNA-directed RNA polymerase (RdRp)) and a structural protein (spike protein (S)) of the virus which are responsible for replication, transcription and host cell recognition. The therapeutic potential of the selected phytochemicals against Covid-19 were also evaluated in comparison with a few commercially available drugs. The binding free energy data suggest that AGP3 could be used as a cost-effective drug-analog for treating covid-19 infection in developing countries.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Natarajan Arul Murugan
- Department of Theoretical Chemistry and Biology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | | |
Collapse
|
210
|
Malato J, Sotzny F, Bauer S, Freitag H, Fonseca A, Grabowska AD, Graça L, Cordeiro C, Nacul L, Lacerda EM, Castro-Marrero J, Scheibenbogen C, Westermeier F, Sepúlveda N. The SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2) in myalgic encephalomyelitis/chronic fatigue syndrome: A meta-analysis of public DNA methylation and gene expression data. Heliyon 2021; 7:e07665. [PMID: 34341773 PMCID: PMC8320404 DOI: 10.1016/j.heliyon.2021.e07665] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/23/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
People with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) often report a high frequency of viral infections and flu-like symptoms during their disease course. Given that this reporting agrees with different immunological abnormalities and altered gene expression profiles observed in the disease, we aimed at answering whether the expression of the human angiotensin-converting enzyme 2 (ACE2), the major cell entry receptor for SARS-CoV-2, is also altered in these patients. In particular, a low expression of ACE2 could be indicative of a high risk of developing COVID-19. We then performed a meta-analysis of public data on CpG DNA methylation and gene expression of this enzyme and its homologous ACE protein in peripheral blood mononuclear cells and related subsets. We found that patients with ME/CFS have decreased methylation levels of four CpG probes in the ACE locus (cg09920557, cg19802564, cg21094739, and cg10468385) and of another probe in the promoter region of the ACE2 gene (cg08559914). We also found a decreased expression of ACE2 but not of ACE in patients when compared to healthy controls. Accordingly, in newly collected data, there was evidence for a significant higher proportion of samples with an ACE2 expression below the limit of detection in patients than healthy controls. Altogether, patients with ME/CFS can be at a higher COVID-19 risk and, if so, they should be considered a priority group for vaccination by public health authorities. To further support this conclusion, similar research is recommended for other human cell entry receptors and cell types, namely, those cells targeted by the virus.
Collapse
Affiliation(s)
- João Malato
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- CEAUL – Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Franziska Sotzny
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Germany
| | - Sandra Bauer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Germany
| | - Helma Freitag
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Germany
| | - André Fonseca
- Faculdade de Ciências e Tecnologia, Universidade do Algarve, Faro, Portugal
| | - Anna D. Grabowska
- Department of Biophysics, Physiology, and Pathophysiology, Medical University of Warsaw, Warsaw, Poland
| | - Luís Graça
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Clara Cordeiro
- CEAUL – Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Ciências e Tecnologia, Universidade do Algarve, Faro, Portugal
| | - Luís Nacul
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Complex Chronic Diseases Program, British Columbia Women's Hospital and Health Centre, Vancouver, British Columbia, Canada
| | - Eliana M. Lacerda
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jesus Castro-Marrero
- Vall d’Hebron Hospital Research Institute, Division of Rheumatology, ME/CFS Unit, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carmen Scheibenbogen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Germany
| | - Francisco Westermeier
- Institute of Biomedical Science, Department of Health Studies, FH Joanneum University of Applied Sciences, Graz, Austria
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O´Higgins, Santiago, Chile
| | - Nuno Sepúlveda
- CEAUL – Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Germany
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
211
|
Lamy PJ, Rébillard X, Vacherot F, de la Taille A. Androgenic hormones and the excess male mortality observed in COVID-19 patients: new convergent data. World J Urol 2021; 39:3121-3123. [PMID: 32488360 PMCID: PMC7266423 DOI: 10.1007/s00345-020-03284-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/27/2020] [Indexed: 12/09/2022] Open
Affiliation(s)
- Pierre-Jean Lamy
- Institut d'Analyse Génomique, IMAGENOME, Labosud-Inovie, Montpellier, France.
| | | | | | - Alexandre de la Taille
- Univ Paris Est Creteil, TRePCa, 94010, Creteil, France
- AP-HP, Hopital Henri Mondor, Service Urology, 94010, Creteil, France
| |
Collapse
|
212
|
Shalash AO, Hussein WM, Skwarczynski M, Toth I. Key Considerations for the Development of Safe and Effective SARS-CoV-2 Subunit Vaccine: A Peptide-Based Vaccine Alternative. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100985. [PMID: 34176237 PMCID: PMC8373118 DOI: 10.1002/advs.202100985] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/18/2021] [Indexed: 05/14/2023]
Abstract
COVID-19 is disastrous to global health and the economy. SARS-CoV-2 infection exhibits similar clinical symptoms and immunopathological sequelae to SARS-CoV infection. Therefore, much of the developmental progress on SARS-CoV vaccines can be utilized for the development of SARS-CoV-2 vaccines. Careful antigen selection during development is always of utmost importance for the production of effective vaccines that do not compromise recipient safety. This holds especially true for SARS-CoV vaccines, as several immunopathological disorders are associated with the activity of structural and nonstructural proteins encoded in the virus's genetic material. Whole viral protein and RNA-encoding full-length proteins contain both protective and "dangerous" sequences, unless pathological fragments are deleted. In light of recent advances, peptide vaccines may present a very safe and effective alternative. Peptide vaccines can avoid immunopathological pro-inflammatory sequences, focus immune responses on neutralizing immunogenic epitopes, avoid off-target antigen loss, combine antigens with different protective roles or mechanisms, even from different viral proteins, and avoid mutant escape by employing highly conserved cryptic epitopes. In this review, an attempt is made to exploit the similarities between SARS-CoV and SARS-CoV-2 in vaccine antigen screening, with particular attention to the pathological and immunogenic properties of SARS proteins.
Collapse
Affiliation(s)
- Ahmed O. Shalash
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt. LuciaQLD4072Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt. LuciaQLD4072Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt. LuciaQLD4072Australia
| | - Istvan Toth
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt. LuciaQLD4072Australia
- Institute for Molecular BioscienceThe University of QueenslandSt. LuciaQLD4072Australia
- School of PharmacyThe University of QueenslandWoolloongabbaQLD4102Australia
| |
Collapse
|
213
|
Poyraz BM, Engin ED, Engin AB, Engin A. The effect of environmental diesel exhaust pollution on SARS-CoV-2 infection: The mechanism of pulmonary ground glass opacity. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 86:103657. [PMID: 33838330 PMCID: PMC8025547 DOI: 10.1016/j.etap.2021.103657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 05/19/2023]
Abstract
Diesel exhaust particles (DEP) are the major components of atmospheric particulate matter (PM) and chronic exposure is recognized to enhance respiratory system complications. Although the spread of SARS-CoV-2 was found to be associated with the PMs, the mechanism by which exposure to DEP increases the risk of SARS-CoV-2 infection is still under discussion. However, diesel fine PM (dPM) elevate the probability of SARS-CoV-2 infection, as it coincides with the increase in the number of ACE2 receptors. Expression of ACE2 and its colocalized activator, transmembrane protease serine 2 (TMPRSS2) facilitate the entry of SARS-CoV-2 into the alveolar epithelial cells exposed to dPM. Thus, the coexistence of PM and SARS-CoV-2 in the environment augments inflammation and exacerbates lung damage. Increased TGF-β1 expression due to DEP accompanies the proliferation of the extracellular matrix. In this case, "multifocal ground-glass opacity" (GGO) in a CT scan is an indication of a cytokine storm and severe pneumonia in COVID-19.
Collapse
Affiliation(s)
| | - Evren Doruk Engin
- Ankara University, Biotechnology Institute, Gumusdere Campus, Kecioren, Ankara, Turkey
| | - Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Ankara, Turkey.
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Ankara, Turkey
| |
Collapse
|
214
|
Widera M, Wilhelm A, Toptan T, Raffel JM, Kowarz E, Roesmann F, Grözinger F, Siemund AL, Luciano V, Külp M, Reis J, Bracharz S, Pallas C, Ciesek S, Marschalek R. Generation of a Sleeping Beauty Transposon-Based Cellular System for Rapid and Sensitive Screening for Compounds and Cellular Factors Limiting SARS-CoV-2 Replication. Front Microbiol 2021; 12:701198. [PMID: 34394046 PMCID: PMC8362758 DOI: 10.3389/fmicb.2021.701198] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the acute respiratory disease COVID-19, which has become a global concern due to its rapid spread. The common methods to monitor and quantitate SARS-CoV-2 infectivity in cell culture are so far time-consuming and labor-intensive. Using the Sleeping Beauty transposase system, we generated a robust and versatile cellular infection model that allows SARS-CoV-2 infection experiments compatible for high-throughput and live cell imaging. The model is based on lung derived A549 cells, which show a profound interferon response and convenient cell culture characteristics. ACE2 and TMPRSS2 were introduced for constitutive expression (A549-AT). Subclones with varying levels of ACE2/TMPRSS2 were screened for optimal SARS-CoV-2 susceptibility. Furthermore, extensive evaluation demonstrated that SARS-CoV-2 infected A549-AT cells were distinguishable from mock-infected cells and already showed approximately 12 h post infection a clear signal to noise ratio in terms of cell roughness, fluorescence and a profound visible cytopathic effect. Moreover, due to the high transfection efficiency and proliferation capacity, Sleeping Beauty transposase-based overexpression cell lines with a second inducible fluorescence reporter cassette (eGFP) can be generated in a very short time, enabling the investigation of host and restriction factors in a doxycycline-inducible manner. Thus, the novel model cell line allows rapid and sensitive monitoring of SARS-CoV-2 infection and the screening for host factors essential for viral replication.
Collapse
Affiliation(s)
- Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt am Main, Goethe University, Frankfurt am Main, Germany
| | - Alexander Wilhelm
- Institute for Medical Virology, University Hospital Frankfurt am Main, Goethe University, Frankfurt am Main, Germany.,Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Tuna Toptan
- Institute for Medical Virology, University Hospital Frankfurt am Main, Goethe University, Frankfurt am Main, Germany
| | - Johanna M Raffel
- Institute for Medical Virology, University Hospital Frankfurt am Main, Goethe University, Frankfurt am Main, Germany
| | - Eric Kowarz
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Fabian Roesmann
- Institute for Medical Virology, University Hospital Frankfurt am Main, Goethe University, Frankfurt am Main, Germany
| | - Finn Grözinger
- Institute for Medical Virology, University Hospital Frankfurt am Main, Goethe University, Frankfurt am Main, Germany
| | - Anna Lena Siemund
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Vanessa Luciano
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Marius Külp
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Jennifer Reis
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Silvia Bracharz
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Christiane Pallas
- Institute for Medical Virology, University Hospital Frankfurt am Main, Goethe University, Frankfurt am Main, Germany
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital Frankfurt am Main, Goethe University, Frankfurt am Main, Germany.,German Center for Infection Research, Braunschweig, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch Translational Medicine and Pharmacology, Frankfurt am Main, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
215
|
Marjomäki V, Kalander K, Hellman M, Permi P. Enteroviruses and coronaviruses: similarities and therapeutic targets. Expert Opin Ther Targets 2021; 25:479-489. [PMID: 34253126 PMCID: PMC8330013 DOI: 10.1080/14728222.2021.1952985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Enteroviruses are common viruses causing a huge number of acute and chronic infections and producing towering economic costs. Similarly, coronaviruses cause seasonal mild infections, epidemics, and even pandemics and can lead to severe respiratory symptoms. It is important to develop broadly acting antiviral molecules to efficiently tackle the infections caused by thes.Areas covered: This review illuminates the differences and similarities between enteroviruses and coronaviruses and examines the most appealing therapeutic targets to combat both virus groups. Publications of both virus groups and deposited structures discovered through PubMed to March 2021 for viral proteases have been evaluated.Expert opinion: The main protease of coronaviruses and enteroviruses share similarities in their structure and function. These proteases process their viral polyproteins and thus drugs that bind to the active site have potential to target both virus groups. It is important to develop drugs that target more evolutionarily conserved processes and proteins. Moreover, it is a wise strategy to concentrate on processes that are similar between several virus families.
Collapse
Affiliation(s)
- Varpu Marjomäki
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Kerttu Kalander
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Maarit Hellman
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Perttu Permi
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
216
|
O Ercelen N, Pekkoc-Uyanik KC, Alpaydin N, Gulay GR, Simsek M. Clinical experience on umbilical cord mesenchymal stem cell treatment in 210 severe and critical COVID-19 cases in Turkey. Stem Cell Rev Rep 2021; 17:1917-1925. [PMID: 34319510 PMCID: PMC8317476 DOI: 10.1007/s12015-021-10214-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 12/17/2022]
Abstract
Objective Treatment for COVID-19 is still urgent need for the critically ill and severe cases. UC-MSC administration has a therapeutic benefit for severe COVID-19 patients even in the recovery period. In this paper, we aimed to present our clinical experience with UC-MSC treatment in severe and critical severe COVID-19 patients. Methods In this study we evaluated the clinical outcome of severe/critically severe 210 COVID-19 patients treated with UC-MSCs, 1–2 × 106 per kilogram to 210 patients from 15/10/2020 until 25/04/2021. Results Out of 99 critically severe intubated patients we have observed good clinical progress/discharged from ICU in 52 (52.5%) patients. Where as 86 (77.5%) of 111 severe unintubated patients discharged from ICU. Intubated 47 (47.5%) patients and unintubated 25 (22.5%) patients pass away. Significantly higher survival was observed in patients who underwent UC-MSCs before intubation (OR = 1.475, 95% CI = 1.193–1.824 p < 0.001). It was observed that the SaO2 parameter tended to improve after UC-MSC therapy compared to all groups. But SaO2 parameter between intubated and unintubated groups was not statistically significant (p > 0.05), while in discharged cases SaO2 parameter was statistically significant (p = 0.01). Besides, there was a statistically significant relation with intubation status, age (OR = 3.868, 95% CI = 0.574–7.152 p = 0.02) and weigh (OR = 6.768, 95% CI = 3.423–10.112 p < 0.001) thus presented an elevated risk for COVID-19. The linear regression analysis confirmed that the high weight was associated with the risk of intubation in COVID-19 (p = 0.001). Conclusions According to our results and from recent studies, UC-MSC treatment is safe with high potential to be used as an added therapeutic treatment for severe COVID-19 patients. Our experience showed that UC-MSC therapy may restore oxygenation and downregulate cytokine storm in patients hospitalized with severe COVID-19. We advice wider randomised studies to discover the detailed therapeutic pathophysiology of the MSCs on COVID-19 patients. Graphical abstract MSCs transplantation improves the damaging effects of the cytokine storm through immunomodulation and improving tissue and organ repair. Severe patients who were unintubated were in the Phase I, while critical patients who were intubated were in the Phase II. The figure is created via biorender application, (BioRender.com).
![]()
Collapse
Affiliation(s)
- Nesrin O Ercelen
- Department of Medical Genetics, Faculty of Medicine, Haliç University, İstanbul, Turkey.
| | | | | | | | - Murat Simsek
- Geneis, Genetic System Solutions, İstanbul, Turkey
| |
Collapse
|
217
|
Sarker J, Das P, Sarker S, Roy AK, Momen AZMR. A Review on Expression, Pathological Roles, and Inhibition of TMPRSS2, the Serine Protease Responsible for SARS-CoV-2 Spike Protein Activation. SCIENTIFICA 2021; 2021:2706789. [PMID: 34336361 PMCID: PMC8313365 DOI: 10.1155/2021/2706789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 05/30/2021] [Accepted: 07/14/2021] [Indexed: 05/08/2023]
Abstract
SARS-CoV-2, the coronavirus responsible for the COVID-19 pandemic, uses the host cell membrane receptor angiotensin-converting enzyme 2 (ACE2) for anchoring its spike protein, and the subsequent membrane fusion process is facilitated by host membrane proteases. Recent studies have shown that transmembrane serine protease 2 (TMPRSS2), a protease known for similar role in previous coronavirus infections, severe acute respiratory syndrome (SARS), and Middle East respiratory syndrome (MERS), is responsible for the proteolytic cleavage of the SARS-CoV-2 spike protein, enabling host cell fusion of the virus. TMPRSS2 is known to be expressed in the epithelial cells of different sites including gastrointestinal, respiratory, and genitourinary system. The infection site of the SARS-CoV-2 correlates with the coexpression sites of ACE2 and TMPRSS2. Besides, age-, sex-, and comorbidity-associated variation in infection rate correlates with the expression rate of TMPRSS2 in those groups. These findings provide valid reasons for the assumption that inhibiting TMPRSS2 can have a beneficial effect in reducing the cellular entry of the virus, ultimately affecting the infection rate and case severity. Several drug development studies are going on to develop potential inhibitors of the protease, using both conventional and computational approaches. Complete understanding of the biological roles of TMPRSS2 is necessary before such therapies are applied.
Collapse
Affiliation(s)
- Jyotirmoy Sarker
- Department of Pharmacy, Jagannath University, Dhaka 1100, Bangladesh
- Department of Pharmacy Systems, Outcomes and Policy, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Pritha Das
- Independent Author, Dhaka 1207, Bangladesh
| | - Sabarni Sarker
- Department of Pharmacy, Jagannath University, Dhaka 1100, Bangladesh
| | - Apurba Kumar Roy
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | | |
Collapse
|
218
|
Gupta VK, Murthy MK, Patil S. Can Host Cell Proteins Like ACE2, ADAM17, TMPRSS2, Androgen Receptor be the Efficient Targets in SARS-CoV-2 Infection? Curr Drug Targets 2021; 22:1149-1157. [PMID: 33243116 DOI: 10.2174/1389450121999201125201112] [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: 07/16/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 11/22/2022]
Abstract
A novel betacoronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV- -2), which caused a large disease outbreak in Wuhan, China in December 2019, is currently spreading across the world. Along with binding of the virus spike with the host cell receptor, fusion of the viral envelope with host cell membranes is a critical step in establishing successful infection of SARS-CoV-2. In this entry process, a diversity of host cell proteases and androgen receptor play a very important role directly or indirectly. These features of SARS-CoV-2 entry contribute to its rapid spread and severe symptoms, high fatality rates among infected patients. This review is based on the latest published literature including review articles, research articles, hypothetical manuscript, preprint articles and official documents. The literature search was made from various published papers on physiological aspects relevant to SARS-CoV and SARS-CoV-2. In this report, we focus on the role of host cell proteases (ACE2, ADAM17, TMPRSS2) and androgen receptor (AR) in SARS-CoV-2 infection. The hypotheses put forth by us are based on the role played by the proteases ACE2, ADAM17, TMPRSS2 and AR in SARS-CoV-2 infection, which were deduced based on various studies. We have also summarized how these host proteins increase the pathology and the infective ability of SARS-CoV-2 and we posit that their inhibition may be a therapeutic option for preventing SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Vivek K Gupta
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra-282004, India
| | - Madhan K Murthy
- Department of Immunology, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra-282004, India
| | - Shripad Patil
- Department of Immunology, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra-282004, India
| |
Collapse
|
219
|
Rawat S, Vrati S, Banerjee A. Neutrophils at the crossroads of acute viral infections and severity. Mol Aspects Med 2021; 81:100996. [PMID: 34284874 PMCID: PMC8286244 DOI: 10.1016/j.mam.2021.100996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022]
Abstract
Neutrophils are versatile immune effector cells essential for mounting a first-line defense against invading pathogens. However, uncontrolled activation can lead to severe life-threatening complications. Neutrophils exist as a heterogeneous population, and their interaction with pathogens and other immune cells may shape the outcome of the host immune response. Diverse classes of viruses, including the recently identified novel SARS-CoV-2, have shown to alter the various aspects of neutrophil biology, offering possibilities for selective intervention. Here, we review heterogeneity within the neutrophil population, highlighting the functional consequences of circulating phenotypes and their critical involvement in exaggerating protective and pathological immune responses against the viruses. We discuss the recent findings of neutrophil extracellular traps (NETs) in COVID-19 pathology and cover other viruses, where neutrophil biology and NETs are crucial for developing disease severity. In the end, we have also pointed out the areas where neutrophil-mediated responses can be finely tuned to outline opportunities for therapeutic manipulation in controlling inflammation against viral infection.
Collapse
Affiliation(s)
- Surender Rawat
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Sudhanshu Vrati
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Arup Banerjee
- Regional Centre for Biotechnology, Faridabad, Haryana, India.
| |
Collapse
|
220
|
Cao W, Feng Q, Wang X. Computational analysis of TMPRSS2 expression in normal and SARS-CoV-2-infected human tissues. Chem Biol Interact 2021; 346:109583. [PMID: 34284028 PMCID: PMC8285370 DOI: 10.1016/j.cbi.2021.109583] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/05/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022]
Abstract
The transmembrane serine protease 2 (TMPRSS2) is a key molecule for SARS-CoV-2 invading human host cells. To provide insights into SARS-CoV-2 infection of various human tissues and understand the potential mechanism of SARS-CoV-2 infection, we investigated TMPRSS2 expression in various normal human tissues and SARS-CoV-2-infected human tissues. Using publicly available datasets, we performed computational analyses of TMPRSS2 expression levels in 30 normal human tissues, and compared them between males and females and between younger (ages ≤ 49 years) and older (ages > 49 years) populations in these tissues. We found that TMPRSS2 expression levels were the highest in the prostate, stomach, pancreas, lungs, small intestine, and salivary gland. The TMPRSS2 protein had relatively high expression levels in the parathyroid gland, stomach, small intestine, pancreas, kidneys, seminal vesicle, epididymis, and prostate. However, TMPRSS2 expression levels were not significantly different between females and males or between younger and older populations in these tissues. The pathways enriched in TMPRSS2-upregulated pan-tissue were mainly involved in immune, metabolism, cell growth and proliferation, stromal signatures, and cancer and other diseases. Many cytokine genes displayed positive expression correlations with TMPRSS2 in pan-tissue, including TNF-α, IL-1, IL-2, IL-4, IL-7, IL-8, IL-12, IL-18, IFN-α, MCP-1, G-CSF, and IP-10. We further analyzed TMPRSS2 expression levels in nasopharyngeal swabs from SARS-CoV-2-infected patients. TMPRSS2 expression levels showed no significant difference between males and females or between younger and older patients. However, they were significantly lower in SARS-CoV-2-infected than in healthy individuals and patients with other viral acute respiratory illnesses. Interestingly, TMPRSS2 expression levels were positively correlated with the enrichment levels of four immune signatures (B cells, CD8+ T cells, NK cells, and interferon response) in SARS-CoV-2-infected patients but likely to be negatively correlated with them in the normal lung tissue. Our data may provide insights into the mechanism of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Wenxiu Cao
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Qiushi Feng
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
221
|
Chazal N. Coronavirus, the King Who Wanted More Than a Crown: From Common to the Highly Pathogenic SARS-CoV-2, Is the Key in the Accessory Genes? Front Microbiol 2021; 12:682603. [PMID: 34335504 PMCID: PMC8317507 DOI: 10.3389/fmicb.2021.682603] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that emerged in late 2019, is the etiologic agent of the current "coronavirus disease 2019" (COVID-19) pandemic, which has serious health implications and a significant global economic impact. Of the seven human coronaviruses, all of which have a zoonotic origin, the pandemic SARS-CoV-2, is the third emerging coronavirus, in the 21st century, highly pathogenic to the human population. Previous human coronavirus outbreaks (SARS-CoV-1 and MERS-CoV) have already provided several valuable information on some of the common molecular and cellular mechanisms of coronavirus infections as well as their origin. However, to meet the new challenge caused by the SARS-CoV-2, a detailed understanding of the biological specificities, as well as knowledge of the origin are crucial to provide information on viral pathogenicity, transmission and epidemiology, and to enable strategies for therapeutic interventions and drug discovery. Therefore, in this review, we summarize the current advances in SARS-CoV-2 knowledges, in light of pre-existing information of other recently emerging coronaviruses. We depict the specificity of the immune response of wild bats and discuss current knowledge of the genetic diversity of bat-hosted coronaviruses that promotes viral genome expansion (accessory gene acquisition). In addition, we describe the basic virology of coronaviruses with a special focus SARS-CoV-2. Finally, we highlight, in detail, the current knowledge of genes and accessory proteins which we postulate to be the major keys to promote virus adaptation to specific hosts (bat and human), to contribute to the suppression of immune responses, as well as to pathogenicity.
Collapse
Affiliation(s)
- Nathalie Chazal
- Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
222
|
Ben-Zuk N, Dechtman ID, Henn I, Weiss L, Afriat A, Krasner E, Gal Y. Potential Prophylactic Treatments for COVID-19. Viruses 2021; 13:1292. [PMID: 34372498 PMCID: PMC8310088 DOI: 10.3390/v13071292] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 01/08/2023] Open
Abstract
The World Health Organization declared the SARS-CoV-2 outbreak a Public Health Emergency of International Concern at the end of January 2020 and a pandemic two months later. The virus primarily spreads between humans via respiratory droplets, and is the causative agent of Coronavirus Disease 2019 (COVID-19), which can vary in severity, from asymptomatic or mild disease (the vast majority of the cases) to respiratory failure, multi-organ failure, and death. Recently, several vaccines were approved for emergency use against SARS-CoV-2. However, their worldwide availability is acutely limited, and therefore, SARS-CoV-2 is still expected to cause significant morbidity and mortality in the upcoming year. Hence, additional countermeasures are needed, particularly pharmaceutical drugs that are widely accessible, safe, scalable, and affordable. In this comprehensive review, we target the prophylactic arena, focusing on small-molecule candidates. In order to consolidate a potential list of such medications, which were categorized as either antivirals, repurposed drugs, or miscellaneous, a thorough screening for relevant clinical trials was conducted. A brief molecular and/or clinical background is provided for each potential drug, rationalizing its prophylactic use as an antiviral or inflammatory modulator. Drug safety profiles are discussed, and current medical indications and research status regarding their relevance to COVID-19 are shortly reviewed. In the near future, a significant body of information regarding the effectiveness of drugs being clinically studied for COVID-19 is expected to accumulate, in addition to information regarding the efficacy of prophylactic treatments.
Collapse
Affiliation(s)
- Noam Ben-Zuk
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
| | - Ido-David Dechtman
- The Israel Defense Force Medical Corps, Tel Hashomer, Military Post 02149, Israel;
- Pulmonology Department, Edith Wolfson Medical Center, 62 Halochamim Street, Holon 5822012, Israel
| | - Itai Henn
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
| | - Libby Weiss
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
| | - Amichay Afriat
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel;
| | - Esther Krasner
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
| | - Yoav Gal
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
- Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| |
Collapse
|
223
|
Raj K, Kaur K, Gupta GD, Singh S. Current understanding on molecular drug targets and emerging treatment strategy for novel coronavirus-19. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:1383-1402. [PMID: 33961065 PMCID: PMC8102151 DOI: 10.1007/s00210-021-02091-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/11/2021] [Indexed: 12/26/2022]
Abstract
SARS-CoV-2 is an enveloped positive-sense RNA virus, contain crown-like spikes on its surface, exceptional of large RNA genome, and a special replication machinery. Common symptoms of SARS-CoV-2 include cough, common cold, fever, sore throat, and a variety of severe acute respiratory disease (SARD) such as pneumonia. SARS-CoV-2 infects epithelial cells, T-cells, macrophages, and dendritic cells and also influences the production and implantation of pro-inflammatory cytokines and chemokines. Repurposing of various drugs during this emergency condition can reduce the rate of mortality as well as time and cost. Two druggable protein and enzyme targets have been selected in this review article due to their crucial role in the viral life cycle. The eukaryotic translation initiation factor (eIF4A), cyclophilin, nucleocapsid protein, spike protein, Angiotensin-converting enzyme 2 (ACE2), 3-chymotrypsin-like cysteine protease (3CLpro), and RNA-dependent RNA polymerase (RdRp) play significant role in early and late phase of SARS-CoV-2 replication and translation. This review paper is based on the rationale of inhibiting of various SARS-CoV-2 proteins and enzymes as novel therapeutic approaches for the management and treatment of patients with SARS-CoV-2 infection. We also discussed the structural and functional relationship of different proteins and enzymes to develop therapeutic approaches for novel coronavirus SARS-CoV-2.
Collapse
Affiliation(s)
- Khadga Raj
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Karamjeet Kaur
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Shamsher Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
224
|
The antiandrogen enzalutamide downregulates TMPRSS2 and reduces cellular entry of SARS-CoV-2 in human lung cells. Nat Commun 2021; 12:4068. [PMID: 34210968 PMCID: PMC8249423 DOI: 10.1038/s41467-021-24342-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 06/09/2021] [Indexed: 12/18/2022] Open
Abstract
SARS-CoV-2 attacks various organs, most destructively the lung, and cellular entry requires two host cell surface proteins: ACE2 and TMPRSS2. Downregulation of one or both of these is thus a potential therapeutic approach for COVID-19. TMPRSS2 is a known target of the androgen receptor, a ligand-activated transcription factor; androgen receptor activation increases TMPRSS2 levels in various tissues, most notably prostate. We show here that treatment with the antiandrogen enzalutamide—a well-tolerated drug widely used in advanced prostate cancer—reduces TMPRSS2 levels in human lung cells and in mouse lung. Importantly, antiandrogens significantly reduced SARS-CoV-2 entry and infection in lung cells. In support of this experimental data, analysis of existing datasets shows striking co-expression of AR and TMPRSS2, including in specific lung cell types targeted by SARS-CoV-2. Together, the data presented provides strong evidence to support clinical trials to assess the efficacy of antiandrogens as a treatment option for COVID-19. TMPRSS2 is regulated by androgen receptor signalling in the prostate, however it is unclear if blocking this signalling is beneficial in the context of SARS-CoV-2 lung infection. Here the authors show that antiandrogen treatment downregulates TMPRSS2 in the lung and reduces viral entry and infection.
Collapse
|
225
|
Barrett CT, Neal HE, Edmonds K, Moncman CL, Thompson R, Branttie JM, Boggs KB, Wu CY, Leung DW, Dutch RE. Effect of clinical isolate or cleavage site mutations in the SARS-CoV-2 spike protein on protein stability, cleavage, and cell-cell fusion. J Biol Chem 2021; 297:100902. [PMID: 34157282 PMCID: PMC8214756 DOI: 10.1016/j.jbc.2021.100902] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 12/14/2022] Open
Abstract
The trimeric severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein (S) is the sole viral protein responsible for both viral binding to a host cell and the membrane fusion event needed for cell entry. In addition to facilitating fusion needed for viral entry, S can also drive cell-cell fusion, a pathogenic effect observed in the lungs of SARS-CoV-2-infected patients. While several studies have investigated S requirements involved in viral particle entry, examination of S stability and factors involved in S cell-cell fusion remain limited. A furin cleavage site at the border between the S1 and S2 subunits (S1/S2) has been identified, along with putative cathepsin L and transmembrane serine protease 2 cleavage sites within S2. We demonstrate that S must be processed at the S1/S2 border in order to mediate cell-cell fusion and that mutations at potential cleavage sites within the S2 subunit alter S processing at the S1/S2 border, thus preventing cell-cell fusion. We also identify residues within the internal fusion peptide and the cytoplasmic tail that modulate S-mediated cell-cell fusion. In addition, we examined S stability and protein cleavage kinetics in a variety of mammalian cell lines, including a bat cell line related to the likely reservoir species for SARS-CoV-2, and provide evidence that proteolytic processing alters the stability of the S trimer. This work therefore offers insight into S stability, proteolytic processing, and factors that mediate S cell-cell fusion, all of which help give a more comprehensive understanding of this high-profile therapeutic target.
Collapse
Affiliation(s)
- Chelsea T Barrett
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Hadley E Neal
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Kearstin Edmonds
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Carole L Moncman
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Rachel Thompson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Jean M Branttie
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Kerri Beth Boggs
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Cheng-Yu Wu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Daisy W Leung
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Rebecca E Dutch
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA.
| |
Collapse
|
226
|
Borges E, Setti AS, Iaconelli A, Braga DPDAF. Current status of the COVID-19 and male reproduction: A review of the literature. Andrology 2021; 9:1066-1075. [PMID: 33998143 PMCID: PMC8222884 DOI: 10.1111/andr.13037] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/09/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), which causes serious respiratory illnesses such as pneumonia and lung failure, was first reported in mid-December 2019 in China and has spread around the world. In addition to causing serious respiratory illnesses such as pneumonia and lung failure, there have been conflicting reports about the presence of SARS-CoV-2 in the semen of patients who were previously diagnosed with COVID-19 and possible implications for the male reproductive tract. OBJECTIVE The goal for the present study was to review the current status of the literature concerning COVID-19 and male reproduction. MATERIAL AND METHODS An electronic literature search was done by using PubMed and Google Scholar databases. Relevant papers, concerning SARS-COV-2 and COVID-19 and male reproduction, published between January 2020 and December 2020 were selected, analyzed and eventually included in the present literature review. RESULTS SARS-CoV-2 may infect any cell type expressing angiotensin-converting enzyme 2 (ACE2), including reproductive cells. Besides the presence of the SARS-CoV-2 receptor, the expression of host proteases, such as transmembrane serine protease 2 (TMPRSS2), is needed to cleave the viral S protein, allowing permanent fusion of the viral and host cell membranes. Here, we aimed to review the current status of the literature concerning COVID-19 and male reproduction. The lack of co-expression of ACE2 and TMPRSS2 in the testis suggests that sperm cells may not be at increased risk of viral entry and spread. However, the presence of orchitis in COVID-19-confirmed patients and compromised sex-related hormonal balance among these patients intrigues reproductive medicine. DISCUSSION SARS-CoV-2 may use alternate receptors to enter certain cell types, or the expression of ACE2 and TMPRSS2 may not be detected in healthy individuals. CONCLUSION COVID-19 challenges all medical areas, including reproductive medicine. It is not yet clear what effects, if any, the COVID-19 pandemic will have on male reproduction. Further research is needed to understand the long-term impact of SARS-CoV-2 on male reproductive function.
Collapse
Affiliation(s)
- Edson Borges
- Fertility Medical GroupSão PauloBrazil
- Instituto Sapientiae – Centro de Estudos e Pesquisa em Reprodução Humana AssistidaSão PauloBrazil
| | - Amanda Souza Setti
- Fertility Medical GroupSão PauloBrazil
- Instituto Sapientiae – Centro de Estudos e Pesquisa em Reprodução Humana AssistidaSão PauloBrazil
| | - Assumpto Iaconelli
- Fertility Medical GroupSão PauloBrazil
- Instituto Sapientiae – Centro de Estudos e Pesquisa em Reprodução Humana AssistidaSão PauloBrazil
| | | |
Collapse
|
227
|
Ma C, Nikiforov A, De Geyter N, Dai X, Morent R, Ostrikov KK. Future antiviral polymers by plasma processing. Prog Polym Sci 2021; 118:101410. [PMID: 33967350 PMCID: PMC8085113 DOI: 10.1016/j.progpolymsci.2021.101410] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/11/2021] [Accepted: 04/22/2021] [Indexed: 12/31/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is largely threatening global public health, social stability, and economy. Efforts of the scientific community are turning to this global crisis and should present future preventative measures. With recent trends in polymer science that use plasma to activate and enhance the functionalities of polymer surfaces by surface etching, surface grafting, coating and activation combined with recent advances in understanding polymer-virus interactions at the nanoscale, it is promising to employ advanced plasma processing for smart antiviral applications. This trend article highlights the innovative and emerging directions and approaches in plasma-based surface engineering to create antiviral polymers. After introducing the unique features of plasma processing of polymers, novel plasma strategies that can be applied to engineer polymers with antiviral properties are presented and critically evaluated. The challenges and future perspectives of exploiting the unique plasma-specific effects to engineer smart polymers with virus-capture, virus-detection, virus-repelling, and/or virus-inactivation functionalities for biomedical applications are analysed and discussed.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- Antiviral polymers
- BSA, bovine serum albumin
- CF4, tetrafluoromethane
- COVID-19, coronavirus disease 2019
- DC, direct current
- H2, hydrogen
- HBV, hepatitis B virus
- HMDSO, hexamethyldisiloxane
- IPNpp, plasma polymerized isopentyl nitrite
- MERS-CoV, middle east respiratory syndrome
- MW, microwave
- NO, nitric oxide
- PC, polycarbonate
- PDMS, polydimethylsiloxane
- PECVD, plasma-enhanced chemical vapour deposition
- PEG, polyethene glycol
- PET, polyethene terephthalate
- PFM, pentafluorophenyl methacrylate
- PP, polypropylene
- PPE, personal protective equipment
- PS, polystyrene
- PTFE, polytetrafluoroethylene
- PVC, polyvinyl chloride
- REF, reference
- RF, radio frequency
- RONS, reactive oxygen and nitrogen species
- RSV, respiratory syncytial virus
- RT-PCR, reverse transcription-polymerase chain reaction
- RV, rhinovirus
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SEM, scanning electron microscopy
- TEOS-O2, tetraethyl orthosilicate and oxygen
- UV, ultraviolet
- WCA, water contact angle
- plasma processing
- surface modification
- ΔD, the variation of the dissipation
- Δf, the frequency shift
Collapse
Affiliation(s)
- Chuanlong Ma
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, B4, 9000 Ghent, Belgium
| | - Anton Nikiforov
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, B4, 9000 Ghent, Belgium
| | - Nathalie De Geyter
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, B4, 9000 Ghent, Belgium
| | - Xiaofeng Dai
- Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Rino Morent
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Sint-Pietersnieuwstraat 41, B4, 9000 Ghent, Belgium
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics and QUT Centre for Materials Science, Queensland University of Technology (QUT), 4000 Brisbane, Australia
| |
Collapse
|
228
|
Mahmoudi S, Balmeh N, Mohammadi N, Sadeghian-Rizi T. The Novel Drug Discovery to Combat COVID-19 by Repressing Important Virus Proteins Involved in Pathogenesis Using Medicinal Herbal Compounds. Avicenna J Med Biotechnol 2021; 13:107-115. [PMID: 34484639 PMCID: PMC8377408 DOI: 10.18502/ajmb.v13i3.6370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The cause of COVID-19 global pandemic is SARS-CoV-2. Given the outbreak of this disease, it is so important to find a treatment. One strategy to cope with COVID-19 is to use the active ingredients of medicinal plants. In this study, the effect of active substances was surveyed in inhibiting four important druggable targets, including S protein, 3CLpro, RdRp, and N protein. RdRp controls the replication of SARS-CoV-2 and is crucial for its life cycle. 3CLpro is the main protease of the virus and could be another therapeutic target. Moreover, N protein and S protein are responsible for SARS-CoV-2 assembly and attaching, respectively. METHODS The 3D structures of herbal active ingredients were prepared and docked with the mentioned SARS-CoV-2 proteins to obtain their affinity. Then, available antiviral drugs introduced in other investigations were docked using similar tools and compared with the results of this study. Finally, other properties of natural compounds were uncovered for drug designing. RESULTS The outcomes of the study revealed that Linarin, Amentoflavone, (-)-Catechin Gallate and Hypericin from Chrysanthemum morifolium, Hypericum perforatum, Humulus lupulus, and Hibiscus sabdariffa had the highest affinity for these basic proteins and in some cases, their affinity was much higher than antiviral medicines. CONCLUSION In addition to having high affinity, these herb active ingredients have antioxidant, vasoprotective, anticarcinogenic, and antiviral properties. Therefore, they can be used as extremely safe therapeutic compounds in drug design studies to control COVID-19.
Collapse
Affiliation(s)
- Samira Mahmoudi
- Department of Microbial Biotechnology, Faculty of Biological Sciences, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Negar Balmeh
- Department of Cell and Molecular Biology, Faculty of Biology, Nour Danesh Institution of Higher Education, Meymeh, Iran
| | - Niloofar Mohammadi
- Department of Sciences and Agricultural Engineering, School of Agricultural Sciences, Pir Bakran Branch, Payame Noor University, Isfahan, Iran
| | - Tahereh Sadeghian-Rizi
- Department of Biotechnology, Faculty of Biological Sciences and Technology, Shahid Ashrafi Esfahani University, Isfahan, Iran
| |
Collapse
|
229
|
Shukla N, Roelle SM, Suzart VG, Bruchez AM, Matreyek KA. Mutants of human ACE2 differentially promote SARS-CoV and SARS-CoV-2 spike mediated infection. PLoS Pathog 2021; 17:e1009715. [PMID: 34270613 PMCID: PMC8284657 DOI: 10.1371/journal.ppat.1009715] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/15/2021] [Indexed: 01/10/2023] Open
Abstract
SARS-CoV and SARS-CoV-2 encode spike proteins that bind human ACE2 on the cell surface to enter target cells during infection. A small fraction of humans encode variants of ACE2, thus altering the biochemical properties at the protein interaction interface. These and other ACE2 coding mutants can reveal how the spike proteins of each virus may differentially engage the ACE2 protein surface during infection. We created an engineered HEK 293T cell line for facile stable transgenic modification, and expressed the major human ACE2 allele or 28 of its missense mutants, 24 of which are possible through single nucleotide changes from the human reference sequence. Infection with SARS-CoV or SARS-CoV-2 spike pseudotyped lentiviruses revealed that high ACE2 cell-surface expression could mask the effects of impaired binding during infection. Drastically reducing ACE2 cell surface expression revealed a range of infection efficiencies across the panel of mutants. Our infection results revealed a non-linear relationship between soluble SARS-CoV-2 RBD binding to ACE2 and pseudovirus infection, supporting a major role for binding avidity during entry. While ACE2 mutants D355N, R357A, and R357T abrogated entry by both SARS-CoV and SARS-CoV-2 spike proteins, the Y41A mutant inhibited SARS-CoV entry much more than SARS-CoV-2, suggesting differential utilization of the ACE2 side-chains within the largely overlapping interaction surfaces utilized by the two CoV spike proteins. These effects correlated well with cytopathic effects observed during SARS-CoV-2 replication in ACE2-mutant cells. The panel of ACE2 mutants also revealed altered ACE2 surface dependencies by the N501Y spike variant, including a near-complete utilization of the K353D ACE2 variant, despite decreased infection mediated by the parental SARS-CoV-2 spike. Our results clarify the relationship between ACE2 abundance, binding, and infection, for various SARS-like coronavirus spike proteins and their mutants, and inform our understanding for how changes to ACE2 sequence may correspond with different susceptibilities to infection.
Collapse
Affiliation(s)
- Nidhi Shukla
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Sarah M. Roelle
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Vinicius G. Suzart
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Anna M. Bruchez
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Kenneth A. Matreyek
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| |
Collapse
|
230
|
Gultom M, Licheri M, Laloli L, Wider M, Strässle M, V'kovski P, Steiner S, Kratzel A, Thao TTN, Probst L, Stalder H, Portmann J, Holwerda M, Ebert N, Stokar-Regenscheit N, Gurtner C, Zanolari P, Posthaus H, Schuller S, Vicente-Santos A, Moreira-Soto A, Corrales-Aguilar E, Ruggli N, Tekes G, von Messling V, Sawatsky B, Thiel V, Dijkman R. Susceptibility of Well-Differentiated Airway Epithelial Cell Cultures from Domestic and Wild Animals to Severe Acute Respiratory Syndrome Coronavirus 2. Emerg Infect Dis 2021; 27:1811-1820. [PMID: 34152956 PMCID: PMC8237902 DOI: 10.3201/eid2707.204660] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread globally, and the number of worldwide cases continues to rise. The zoonotic origins of SARS-CoV-2 and its intermediate and potential spillback host reservoirs, besides humans, remain largely unknown. Because of ethical and experimental constraints and more important, to reduce and refine animal experimentation, we used our repository of well-differentiated airway epithelial cell (AEC) cultures from various domesticated and wildlife animal species to assess their susceptibility to SARS-CoV-2. We observed that SARS-CoV-2 replicated efficiently only in monkey and cat AEC culture models. Whole-genome sequencing of progeny viruses revealed no obvious signs of nucleotide transitions required for SARS-CoV-2 to productively infect monkey and cat AEC cultures. Our findings, together with previous reports of human-to-animal spillover events, warrant close surveillance to determine the potential role of cats, monkeys, and closely related species as spillback reservoirs for SARS-CoV-2.
Collapse
|
231
|
Identification of a High-Frequency Intrahost SARS-CoV-2 Spike Variant with Enhanced Cytopathic and Fusogenic Effects. mBio 2021; 12:e0078821. [PMID: 34182784 PMCID: PMC8262852 DOI: 10.1128/mbio.00788-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a virus that is continuously evolving. Although its RNA-dependent RNA polymerase exhibits some exonuclease proofreading activity, viral sequence diversity can be produced by replication errors and host factors. A diversity of genetic variants can be observed in the intrahost viral population structure of infected individuals. Most mutations will follow a neutral molecular evolution and will not make significant contributions to variations within and between infected hosts. Herein, we profiled the intrasample genetic diversity of SARS-CoV-2 variants, also known as quasispecies, using high-throughput sequencing data sets from 15,289 infected individuals and infected cell lines. Despite high mutational background, we identified recurrent intragenetic variable positions in the samples analyzed, including several positions at the end of the gene encoding the viral spike (S) protein. Strikingly, we observed a high frequency of C→A missense mutations resulting in the S protein lacking the last 20 amino acids (SΔ20). We found that this truncated S protein undergoes increased processing and increased syncytium formation, presumably due to escaping M protein retention in intracellular compartments. Our findings suggest the emergence of a high-frequency viral sublineage that is not horizontally transmitted but potentially involved in intrahost disease cytopathic effects. IMPORTANCE The mutation rate and evolution of RNA viruses correlate with viral adaptation. While most mutations do not make significant contributions to viral molecular evolution, some are naturally selected and produce variants through positive selection. Many SARS-CoV-2 variants have been recently described and show phenotypic selection toward more infectious viruses. Our study describes another type of variant that does not contribute to interhost heterogeneity but rather phenotypic selection toward variants that might have increased cytopathic effects. We identified that a C-terminal truncation of the spike protein removes an important endoplasmic reticulum (ER) retention signal, which consequently results in a spike variant that easily travels through the Golgi complex toward the plasma membrane in a preactivated conformation, leading to increased syncytium formation.
Collapse
|
232
|
Bohan D, Ert HV, Ruggio N, Rogers KJ, Badreddine M, Aguilar Briseño JA, Rojas Chavez RA, Gao B, Stokowy T, Christakou E, Micklem D, Gausdal G, Haim H, Minna J, Lorens JB, Maury W. Phosphatidylserine Receptors Enhance SARS-CoV-2 Infection: AXL as a Therapeutic Target for COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34159331 PMCID: PMC8219095 DOI: 10.1101/2021.06.15.448419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Phosphatidylserine (PS) receptors are PS binding proteins that mediate uptake of apoptotic bodies. Many enveloped viruses utilize this PS/PS receptor mechanism to adhere to and internalize into the endosomal compartment of cells and this is termed apoptotic mimicry. For viruses that have a mechanism(s) of endosomal escape, apoptotic mimicry is a productive route of virus entry. We evaluated if PS receptors serve as cell surface receptors for SARS-CoV-2 and found that the PS receptors, AXL, TIM-1 and TIM-4, facilitated virus infection when low concentrations of the SARS-CoV-2 cognate receptor, ACE2, was present. Consistent with the established mechanism of PS receptor utilization by other viruses, PS liposomes competed with SARS-CoV-2 for binding and entry. We demonstrated that this PS receptor enhances SARS-CoV-2 binding to and infection of an array of human lung cell lines and is an under-appreciated but potentially important host factor facilitating SARS-CoV-2 entry.
Collapse
Affiliation(s)
- Dana Bohan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| | - Hanora Van Ert
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| | - Natalie Ruggio
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| | - Kai J Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| | - Mohammad Badreddine
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| | | | | | - Boning Gao
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - Tomasz Stokowy
- Department of Biomedicine, University of Bergen, Bergen Norway
| | - Eleni Christakou
- Department of Biomedicine, University of Bergen, Bergen Norway.,BerGenBio ASA, Bergen, Norway
| | | | | | - Hillel Haim
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| | - John Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - James B Lorens
- Department of Biomedicine, University of Bergen, Bergen Norway
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA
| |
Collapse
|
233
|
Structural basis of covalent inhibitory mechanism of TMPRSS2-related serine proteases by camostat. J Virol 2021; 95:e0086121. [PMID: 34160253 PMCID: PMC8428381 DOI: 10.1128/jvi.00861-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the viral pathogen causing the coronavirus disease 2019 (COVID-19) global pandemic. No effective treatment for COVID-19 has been established yet. The serine protease transmembrane protease serine 2 (TMPRSS2) is essential for viral spread and pathogenicity by facilitating the entry of SARS-CoV-2 into host cells. The protease inhibitor camostat, an anticoagulant used in the clinic, has potential anti-inflammatory and antiviral activities against COVID-19. However, the potential mechanisms of viral resistance and antiviral activity of camostat are unclear. Herein, we demonstrate high inhibitory potencies of camostat for a panel of serine proteases, indicating that camostat is a broad-spectrum inhibitor of serine proteases. In addition, we determined the crystal structure of camostat in complex with a serine protease (uPA [urokinase-type plasminogen activator]), which reveals that camostat is inserted in the S1 pocket of uPA but is hydrolyzed by uPA, and the cleaved camostat covalently binds to Ser195. We also generated a homology model of the structure of the TMPRSS2 serine protease domain. The model shows that camostat uses the same inhibitory mechanism to inhibit the activity of TMPRSS2, subsequently preventing SARS-CoV-2 spread. IMPORTANCE Serine proteases are a large family of enzymes critical for multiple physiological processes and proven diagnostic and therapeutic targets in several clinical indications. The serine protease transmembrane protease serine 2 (TMPRSS2) was recently found to mediate SARS-CoV-2 entry into the host. Camostat mesylate (FOY 305), a serine protease inhibitor active against TMPRSS2 and used for the treatment of oral squamous cell carcinoma and chronic pancreatitis, inhibits SARS-CoV-2 infection of human lung cells. However, the direct inhibition mechanism of camostat mesylate for TMPRSS2 is unclear. Herein, we demonstrate that camostat uses the same inhibitory mechanism to inhibit the activity of TMPRSS2 as uPA, subsequently preventing SARS-CoV-2 spread.
Collapse
|
234
|
Hussman JP. Severe Clinical Worsening in COVID-19 and Potential Mechanisms of Immune-Enhanced Disease. Front Med (Lausanne) 2021; 8:637642. [PMID: 34239884 PMCID: PMC8258105 DOI: 10.3389/fmed.2021.637642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Infection by the novel SARS-CoV-2 coronavirus produces a range of outcomes, with the majority of cases producing mild or asymptomatic effects, and a smaller subset progressing to critical or fatal COVID-19 disease featuring severe acute respiratory distress. Although the mechanisms driving severe disease progression remain unknown, it is possible that the abrupt clinical deterioration observed in patients with critical disease corresponds to a discrete underlying expansion of viral tropism, from infection of cells comprising respiratory linings and alveolar epithelia to direct infection and activation of inflammatory monocytes and macrophages. Dysregulated immune responses could then contribute to disease severity. This article discusses the potential role of monocyte/macrophage (Mo/Mϕ) infection by SARS-CoV-2 in mediating the immune response in severe COVID-19. Additional mechanisms of immune-enhanced disease, comprising maladaptive immune responses that may aggravate rather than alleviate severity, are also discussed. Severe acute clinical worsening in COVID-19 patients may be influenced by the emergence of antibodies that participate in hyperinflammatory monocyte response, release of neutrophil extracellular traps (NETs), thrombosis, platelet apoptosis, viral entry into Fc gamma receptor (FcγR)-expressing immune cells, and induction of autoantibodies with cross-reactivity against host proteins. While the potential roles of Mo/Mϕ infection and immune-enhanced pathology in COVID-19 are consistent with a broad range of clinical and laboratory findings, their prominence remains tentative pending further validation. In the interim, these proposed mechanisms present immediate avenues of inquiry that may help to evaluate the safety of candidate vaccines and antibody-based therapeutics, and to support consideration of pathway-informed, well-tolerated therapeutic candidates targeting the dysregulated immune response.
Collapse
|
235
|
Najafi Fard S, Petrone L, Petruccioli E, Alonzi T, Matusali G, Colavita F, Castilletti C, Capobianchi MR, Goletti D. In Vitro Models for Studying Entry, Tissue Tropism, and Therapeutic Approaches of Highly Pathogenic Coronaviruses. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8856018. [PMID: 34239932 PMCID: PMC8221881 DOI: 10.1155/2021/8856018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/27/2021] [Accepted: 06/05/2021] [Indexed: 12/31/2022]
Abstract
Coronaviruses (CoVs) are enveloped nonsegmented positive-sense RNA viruses belonging to the family Coronaviridae that contain the largest genome among RNA viruses. Their genome encodes 4 major structural proteins, and among them, the Spike (S) protein plays a crucial role in determining the viral tropism. It mediates viral attachment to the host cell, fusion to the membranes, and cell entry using cellular proteases as activators. Several in vitro models have been developed to study the CoVs entry, pathogenesis, and possible therapeutic approaches. This article is aimed at summarizing the current knowledge about the use of relevant methodologies and cell lines permissive for CoV life cycle studies. The synthesis of this information can be useful for setting up specific experimental procedures. We also discuss different strategies for inhibiting the binding of the S protein to the cell receptors and the fusion process which may offer opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Saeid Najafi Fard
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Linda Petrone
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Elisa Petruccioli
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Tonino Alonzi
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Giulia Matusali
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Francesca Colavita
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| | - Delia Goletti
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy
| |
Collapse
|
236
|
Zhang Q, Xiang R, Huo S, Zhou Y, Jiang S, Wang Q, Yu F. Molecular mechanism of interaction between SARS-CoV-2 and host cells and interventional therapy. Signal Transduct Target Ther 2021; 6:233. [PMID: 34117216 PMCID: PMC8193598 DOI: 10.1038/s41392-021-00653-w] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 02/05/2023] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has resulted in an unprecedented setback for global economy and health. SARS-CoV-2 has an exceptionally high level of transmissibility and extremely broad tissue tropism. However, the underlying molecular mechanism responsible for sustaining this degree of virulence remains largely unexplored. In this article, we review the current knowledge and crucial information about how SARS-CoV-2 attaches on the surface of host cells through a variety of receptors, such as ACE2, neuropilin-1, AXL, and antibody-FcγR complexes. We further explain how its spike (S) protein undergoes conformational transition from prefusion to postfusion with the help of proteases like furin, TMPRSS2, and cathepsins. We then review the ongoing experimental studies and clinical trials of antibodies, peptides, or small-molecule compounds with anti-SARS-CoV-2 activity, and discuss how these antiviral therapies targeting host-pathogen interaction could potentially suppress viral attachment, reduce the exposure of fusion peptide to curtail membrane fusion and block the formation of six-helix bundle (6-HB) fusion core. Finally, the specter of rapidly emerging SARS-CoV-2 variants deserves a serious review of broad-spectrum drugs or vaccines for long-term prevention and control of COVID-19 in the future.
Collapse
Affiliation(s)
- Qianqian Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Rong Xiang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yunjiao Zhou
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Fei Yu
- College of Life Sciences, Hebei Agricultural University, Baoding, China.
| |
Collapse
|
237
|
To Trap a Pathogen: Neutrophil Extracellular Traps and Their Role in Mucosal Epithelial and Skin Diseases. Cells 2021; 10:cells10061469. [PMID: 34208037 PMCID: PMC8230648 DOI: 10.3390/cells10061469] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/18/2022] Open
Abstract
Neutrophils are the most abundant circulating innate immune cells and comprise the first immune defense line, as they are the most rapidly recruited cells at sites of infection or inflammation. Their main microbicidal mechanisms are degranulation, phagocytosis, cytokine secretion and the formation of extracellular traps. Neutrophil extracellular traps (NETs) are a microbicidal mechanism that involves neutrophil death. Since their discovery, in vitro and in vivo neutrophils have been challenged with a range of stimuli capable of inducing or inhibiting NET formation, with the objective to understand its function and regulation in health and disease. These networks composed of DNA and granular components are capable of immobilizing and killing pathogens. They comprise enzymes such as myeloperoxidase, elastase, cathepsin G, acid hydrolases and cationic peptides, all with antimicrobial and antifungal activity. Therefore, the excessive formation of NETs can also lead to tissue damage and promote local and systemic inflammation. Based on this concept, in this review, we focus on the role of NETs in different infectious and inflammatory diseases of the mucosal epithelia and skin.
Collapse
|
238
|
Tavakol S, Alavijeh MS, Seifalian AM. COVID-19 Vaccines in Clinical Trials and their Mode of Action for Immunity against the Virus. Curr Pharm Des 2021; 27:1553-1563. [PMID: 33100195 DOI: 10.2174/1381612826666201023143956] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/10/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
For nearly two decades, coronaviruses have caused many health and economic problems, while no effective commercial vaccine has yet been developed. It is worth mentioning that despite some mutations and recombination in SARS-CoV-2, its genotype is very close to the original strain from Wuhan, China. Therefore, the development of an effective vaccine would be promising. It might be hypothesized that BCG vaccination is performed in high-risk populations before the commercialization of an effective SARS-CoV-2 vaccine. However, the development of an effective vaccine without considering the adverse immune reactions derived from antibody-dependent or cell-based immune enhancement may threaten vaccinated people's lives and long-term side effects must be considered. To this end, targeting of the receptor-binding domain (RBD) in spike and not whole spike, glycolization of FC receptors, PD-1 blockers, CPPs, etc., are promising. Therefore, the subunit vaccines or RNA vaccines that encode the RBP segment of the spike are of interest. To enhance the vaccine efficacy, its co-delivery with an adjuvant has been recommended. Nanoparticles modulate immune response with higher efficiency than the soluble form of antigens and can be functionalized with the positively charged moieties and ligands of targeted cells, such as dendritic cells, to increase cellular uptake of the antigens and their presentation on the surface of immune cells. This research aimed to discuss the COVID-19 vaccines entering the clinical trial and their mode of action effective immunity against the virus and discusses their advantages compared to each other.
Collapse
Affiliation(s)
- Shima Tavakol
- Pharmidex Pharmaceutical Services Ltd., London, United Kingdom
| | - Mo S Alavijeh
- Pharmidex Pharmaceutical Services Ltd., London, United Kingdom
| | - Alexander M Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
239
|
Khodajou-Masouleh H, Shahangian SS, Rasti B. Reinforcing our defense or weakening the enemy? A comparative overview of defensive and offensive strategies developed to confront COVID-19. Drug Metab Rev 2021; 53:508-541. [PMID: 33980089 DOI: 10.1080/03602532.2021.1928686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Developing effective strategies to confront coronavirus disease 2019 (COVID-19) has become one of the greatest concerns of the scientific community. In addition to the vast number of global mortalities due to COVID-19, since its outbreak, almost every aspect of human lives has changed one way or another. In the present review, various defensive and offensive strategies developed to confront COVID-19 are illustrated. The Administration of immune-boosting micronutrients/agents, as well as the inhibition of the activity of incompetent gatekeepers, including some host cell receptors (e.g. ACE2) and proteases (e.g. TMPRSS2), are some efficient defensive strategies. Antibody/phage therapies and specifically vaccines also play a prominent role in the enhancement of host defense against COVID-19. Nanotechnology, however, can considerably weaken the virulence of SARS-CoV-2, utilizing fake cellular locks (compounds mimicking cell receptors) to block the viral keys (spike proteins). Generally, two strategies are developed to interfere with the binding of spike proteins to the host cell receptors, either utilizing fake cellular locks to block the viral keys or utilizing fake viral keys to block the cellular locks. Due to their evolutionary conserved nature, viral enzymes, including 3CLpro, PLpro, RdRp, and helicase are highly potential targets for drug repurposing strategy. Thus, various steps of viral replication/transcription can effectively be blocked by their inhibition, leading to the elimination of SARS-CoV-2. Moreover, RNA decoy and CRISPR technologies likely offer the best offensive strategies after viral entry into the host cells, inhibiting the viral replication/assembly in the infected cells and substantially reducing the quantity of viral progeny.
Collapse
Affiliation(s)
| | - S Shirin Shahangian
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Behnam Rasti
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| |
Collapse
|
240
|
Raghav PK, Kalyanaraman K, Kumar D. Human cell receptors: potential drug targets to combat COVID-19. Amino Acids 2021; 53:813-842. [PMID: 33950300 PMCID: PMC8097256 DOI: 10.1007/s00726-021-02991-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 04/21/2021] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the coronavirus disease 2019 (COVID-19). The World Health Organization (WHO) has announced that COVID-19 is a pandemic having a higher spread rate rather than the mortality. Identification of a potential approach or therapy against COVID-19 is still under consideration. Therefore, it is essential to have an insight into SARS-CoV-2, its interacting partner, and domains for an effective treatment. The present study is divided into three main categories, including SARS-CoV-2 prominent receptor and its expression levels, other interacting partners, and their binding domains. The first section focuses primarily on coronaviruses' general aspects (SARS-CoV-2, SARS-CoV, and the Middle East Respiratory Syndrome Coronaviruses (MERS-CoV)) their structures, similarities, and mode of infections. The second section discusses the host receptors which includes the human targets of coronaviruses like dipeptidyl peptidase 4 (DPP4), CD147, CD209L, Angiotensin-Converting Enzyme 2 (ACE2), and other miscellaneous targets (type-II transmembrane serine proteases (TTSPs), furin, trypsin, cathepsins, thermolysin, elastase, phosphatidylinositol 3-phosphate 5-kinase, two-pore segment channel, and epithelium sodium channel C-α subunit). The human cell receptor, ACE2 plays an essential role in the Renin-Angiotensin system (RAS) pathway and COVID-19. Thus, this section also discusses the ACE2 expression and risk of COVID-19 infectivity in various organs and tissues such as the liver, lungs, intestine, heart, and reproductive system in the human body. Absence of ACE2 protein expression in immune cells could be used for limiting the SARS-CoV-2 infection. The third section covers the current available approaches for COVID-19 treatment. Overall, this review focuses on the critical role of human cell receptors involved in coronavirus pathogenesis, which would likely be used in designing target-specific drugs to combat COVID-19.
Collapse
Affiliation(s)
| | - Keerthana Kalyanaraman
- Amity Institute of Biotechnology, Amity University, Sector-125, Noida, Uttar Pradesh, India
| | - Dinesh Kumar
- ICMR-National Institute of Cancer Prevention & Research, Noida, 201301, India.
| |
Collapse
|
241
|
Soumya RS, Unni TG, Raghu KG. Impact of COVID-19 on the Cardiovascular System: A Review of Available Reports. Cardiovasc Drugs Ther 2021; 35:411-425. [PMID: 32926272 PMCID: PMC7487338 DOI: 10.1007/s10557-020-07073-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/01/2020] [Indexed: 01/08/2023]
Abstract
The recent emergence of the coronavirus disease 19 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in China is now a global health emergency. The transmission of SARS-CoV-2 is mainly via human-to-human contact. This virus is expected to be of zoonotic origin and has a high genome identity to that of bat derived SARS-like coronavirus. Various stringent measures have been implemented to lower person-to-person transmission of COVID-19. Particular observations and attempts have been made to reduce transmission in vulnerable populations, including older adults, children, and healthcare providers. This novel CoV enters the cells through the angiotensin-converting enzyme 2 (ACE2) receptor. There is a higher risk of COVID-19 infection among those with preexisting cardiovascular diseases (CVD), and it has been connected with various direct and indirect complications, including myocarditis, acute myocardial injury, venous thromboembolism, and arrhythmias. This article summarizes the various cardiovascular complications and mechanisms responsible for the same with COVID-19 infection. For the benefit of the scientific community and public, the effect of COVID-19 on major vital organs such as the kidneys, liver, and intestines has been briefly discussed. In this review, we also discuss drugs in different stages of clinical trials and their associated complications, as well as the details of vaccines in various stages of development.
Collapse
Affiliation(s)
- R S Soumya
- Biochemistry and Molecular Mechanism Laboratory, Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, 695019, India
| | - T Govindan Unni
- Department of Cardiology, Jubilee Mission Medical College & Research Centre, Thrissur, Kerala, 680005, India
| | - K G Raghu
- Biochemistry and Molecular Mechanism Laboratory, Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, 695019, India.
| |
Collapse
|
242
|
Braga L, Ali H, Secco I, Chiavacci E, Neves G, Goldhill D, Penn R, Jimenez-Guardeño JM, Ortega-Prieto AM, Bussani R, Cannatà A, Rizzari G, Collesi C, Schneider E, Arosio D, Shah AM, Barclay WS, Malim MH, Burrone J, Giacca M. Drugs that inhibit TMEM16 proteins block SARS-CoV-2 spike-induced syncytia. Nature 2021; 594:88-93. [PMID: 33827113 PMCID: PMC7611055 DOI: 10.1038/s41586-021-03491-6] [Citation(s) in RCA: 282] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 03/25/2021] [Indexed: 02/01/2023]
Abstract
COVID-19 is a disease with unique characteristics that include lung thrombosis1, frequent diarrhoea2, abnormal activation of the inflammatory response3 and rapid deterioration of lung function consistent with alveolar oedema4. The pathological substrate for these findings remains unknown. Here we show that the lungs of patients with COVID-19 contain infected pneumocytes with abnormal morphology and frequent multinucleation. The generation of these syncytia results from activation of the SARS-CoV-2 spike protein at the cell plasma membrane level. On the basis of these observations, we performed two high-content microscopy-based screenings with more than 3,000 approved drugs to search for inhibitors of spike-driven syncytia. We converged on the identification of 83 drugs that inhibited spike-mediated cell fusion, several of which belonged to defined pharmacological classes. We focused our attention on effective drugs that also protected against virus replication and associated cytopathicity. One of the most effective molecules was the antihelminthic drug niclosamide, which markedly blunted calcium oscillations and membrane conductance in spike-expressing cells by suppressing the activity of TMEM16F (also known as anoctamin 6), a calcium-activated ion channel and scramblase that is responsible for exposure of phosphatidylserine on the cell surface. These findings suggest a potential mechanism for COVID-19 disease pathogenesis and support the repurposing of niclosamide for therapy.
Collapse
Affiliation(s)
- Luca Braga
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Hashim Ali
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Ilaria Secco
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Elena Chiavacci
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Guilherme Neves
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daniel Goldhill
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Rebecca Penn
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Jose M Jimenez-Guardeño
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Ana M Ortega-Prieto
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Rossana Bussani
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Antonio Cannatà
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Giorgia Rizzari
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Chiara Collesi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Edoardo Schneider
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Daniele Arosio
- Istituto di Biofisica (IBF), Consiglio Nazionale delle Ricerche (CNR), Trento, Italy
| | - Ajay M Shah
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Wendy S Barclay
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Michael H Malim
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Juan Burrone
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Mauro Giacca
- King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK.
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.
| |
Collapse
|
243
|
Carroll EL, Bailo M, Reihill JA, Crilly A, Lockhart JC, Litherland GJ, Lundy FT, McGarvey LP, Hollywood MA, Martin SL. Trypsin-Like Proteases and Their Role in Muco-Obstructive Lung Diseases. Int J Mol Sci 2021; 22:5817. [PMID: 34072295 PMCID: PMC8199346 DOI: 10.3390/ijms22115817] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Trypsin-like proteases (TLPs) belong to a family of serine enzymes with primary substrate specificities for the basic residues, lysine and arginine, in the P1 position. Whilst initially perceived as soluble enzymes that are extracellularly secreted, a number of novel TLPs that are anchored in the cell membrane have since been discovered. Muco-obstructive lung diseases (MucOLDs) are characterised by the accumulation of hyper-concentrated mucus in the small airways, leading to persistent inflammation, infection and dysregulated protease activity. Although neutrophilic serine proteases, particularly neutrophil elastase, have been implicated in the propagation of inflammation and local tissue destruction, it is likely that the serine TLPs also contribute to various disease-relevant processes given the roles that a number of these enzymes play in the activation of both the epithelial sodium channel (ENaC) and protease-activated receptor 2 (PAR2). More recently, significant attention has focused on the activation of viruses such as SARS-CoV-2 by host TLPs. The purpose of this review was to highlight key TLPs linked to the activation of ENaC and PAR2 and their association with airway dehydration and inflammatory signalling pathways, respectively. The role of TLPs in viral infectivity will also be discussed in the context of the inhibition of TLP activities and the potential of these proteases as therapeutic targets.
Collapse
Affiliation(s)
- Emma L. Carroll
- School of Pharmacy, Queen’s University, Belfast BT9 7BL, UK; (E.L.C.); (J.A.R.)
| | - Mariarca Bailo
- Institute for Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK; (M.B.); (A.C.); (J.C.L.); (G.J.L.)
| | - James A. Reihill
- School of Pharmacy, Queen’s University, Belfast BT9 7BL, UK; (E.L.C.); (J.A.R.)
| | - Anne Crilly
- Institute for Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK; (M.B.); (A.C.); (J.C.L.); (G.J.L.)
| | - John C. Lockhart
- Institute for Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK; (M.B.); (A.C.); (J.C.L.); (G.J.L.)
| | - Gary J. Litherland
- Institute for Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK; (M.B.); (A.C.); (J.C.L.); (G.J.L.)
| | - Fionnuala T. Lundy
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University, Belfast BT9 7BL, UK; (F.T.L.); (L.P.M.)
| | - Lorcan P. McGarvey
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University, Belfast BT9 7BL, UK; (F.T.L.); (L.P.M.)
| | - Mark A. Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology, A91 HRK2 Dundalk, Ireland;
| | - S. Lorraine Martin
- School of Pharmacy, Queen’s University, Belfast BT9 7BL, UK; (E.L.C.); (J.A.R.)
| |
Collapse
|
244
|
Role of host factors in SARS-CoV-2 entry. J Biol Chem 2021; 297:100847. [PMID: 34058196 PMCID: PMC8160279 DOI: 10.1016/j.jbc.2021.100847] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
The zoonotic transmission of highly pathogenic coronaviruses into the human population is a pressing concern highlighted by the ongoing SARS-CoV-2 pandemic. Recent work has helped to illuminate much about the mechanisms of SARS-CoV-2 entry into the cell, which determines host- and tissue-specific tropism, pathogenicity, and zoonotic transmission. Here we discuss current findings on the factors governing SARS-CoV-2 entry. We first reviewed key features of the viral spike protein (S) mediating fusion of the viral envelope and host cell membrane through binding to the SARS-CoV-2 receptor, angiotensin-converting enzyme 2. We then examined the roles of host proteases including transmembrane protease serine 2 and cathepsins in processing S for virus entry and the impact of this processing on endosomal and plasma membrane virus entry routes. We further discussed recent work on several host cofactors that enhance SARS-CoV-2 entry including Neuropilin-1, CD147, phosphatidylserine receptors, heparan sulfate proteoglycans, sialic acids, and C-type lectins. Finally, we discussed two key host restriction factors, i.e., interferon-induced transmembrane proteins and lymphocyte antigen 6 complex locus E, which can disrupt SARS-CoV-2 entry. The features of SARS-CoV-2 are presented in the context of other human coronaviruses, highlighting unique aspects. In addition, we identify the gaps in understanding of SARS-CoV-2 entry that will need to be addressed by future studies.
Collapse
|
245
|
Rebendenne A, Roy P, Bonaventure B, Chaves VAL, Desmarets L, Rouillé Y, Tauziet M, Arnaud-Arnould M, Giovannini D, Lee Y, DeWeirdt P, Hegde M, Garcia de GF, McKellar J, Wencker M, Dubuisson J, Belouzard S, Moncorgé O, Doench JG, Goujon C. Bidirectional genome-wide CRISPR screens reveal host factors regulating SARS-CoV-2, MERS-CoV and seasonal HCoVs. RESEARCH SQUARE 2021:rs.3.rs-555275. [PMID: 34075371 PMCID: PMC8168385 DOI: 10.21203/rs.3.rs-555275/v1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Several genome-wide CRISPR knockout screens have been conducted to identify host factors regulating SARS-CoV-2 replication, but the models used have often relied on overexpression of ACE2 receptor. Additionally, such screens have yet to identify the protease TMPRSS2, known to be important for viral entry at the plasma membrane. Here, we conducted a meta-analysis of these screens and showed a high level of cell-type specificity of the identified hits, arguing for the necessity of additional models to uncover the full landscape of SARS-CoV-2 host factors. We performed genome-wide knockout and activation CRISPR screens in Calu-3 lung epithelial cells, as well as knockout screens in Caco-2 intestinal cells. In addition to identifying ACE2 and TMPRSS2 as top hits, our study reveals a series of so far unidentified and critical host-dependency factors, including the Adaptins AP1G1 and AP1B1 and the flippase ATP8B1. Moreover, new anti-SARS-CoV-2 proteins with potent activity, including several membrane-associated Mucins, IL6R, and CD44 were identified. We further observed that these genes mostly acted at the critical step of viral entry, with the notable exception of ATP8B1, the knockout of which prevented late stages of viral replication. Exploring the pro- and anti-viral breadth of these genes using highly pathogenic MERS-CoV, seasonal HCoV-NL63 and -229E and influenza A orthomyxovirus, we reveal that some genes such as AP1G1 and ATP8B1 are general coronavirus cofactors. In contrast, Mucins recapitulated their known role as a general antiviral defense mechanism. These results demonstrate the value of considering multiple cell models and perturbational modalities for understanding SARS-CoV-2 replication and provide a list of potential new targets for therapeutic interventions.
Collapse
Affiliation(s)
| | - Priyanka Roy
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | | | | | - Lowiese Desmarets
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, France
| | - Yves Rouillé
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, France
| | | | | | | | - Yenarae Lee
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Peter DeWeirdt
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Mudra Hegde
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | | | | | | | - Jean Dubuisson
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, France
| | - Sandrine Belouzard
- Lille University, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, France
| | | | - John G. Doench
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, USA
| | | |
Collapse
|
246
|
Shahriari Felordi M, Memarnejadian A, Najimi M, Vosough M. Is There any Alternative Receptor for SARS-CoV-2? CELL JOURNAL 2021; 23:247-250. [PMID: 34096226 PMCID: PMC8181318 DOI: 10.22074/cellj.2021.7977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/17/2021] [Indexed: 12/15/2022]
Abstract
Angiotensin-converting enzyme II (ACE2) in association with type II transmembrane serine protease (TMPRSS2) is
considered the main receptor of SARS-CoV-2. However, considering the clinical complications of COVID-19 in different
organs, there is no strong association between the abundance of ACE2/TMPRSS2 co-expression and clinical features
of the disease and the severity of complications. Since SARS-CoV-2 affects certain organs that lack or have low
expression of ACE2/TMPRSS2, it may be possible that the virus employs other receptors for colonization and entry.
Based on recent studies, glucose-regulated protein 78 (GRP78) can be a potential alternative receptor for SARS-CoV-2
entry. In this letter, supporting evidence proposed GRP78 as an alternative receptor in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mahtab Shahriari Felordi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
247
|
Müller P, Maus H, Hammerschmidt SJ, Knaff P, Mailänder V, Schirmeister T, Kersten C. Interfering with Host Proteases in SARS-CoV-2 Entry as a Promising Therapeutic Strategy. Curr Med Chem 2021; 29:635-665. [PMID: 34042026 DOI: 10.2174/0929867328666210526111318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 01/10/2023]
Abstract
Due to its fast international spread and substantial mortality, the coronavirus disease COVID-19 evolved to a global threat. Since currently, there is no causative drug against this viral infection available, science is striving for new drugs and approaches to treat the new disease. Studies have shown that the cell entry of coronaviruses into host cells takes place through the binding of the viral spike (S) protein to cell receptors. Priming of the S protein occurs via hydrolysis by different host proteases. The inhibition of these proteases could impair the processing of the S protein, thereby affecting the interaction with the host-cell receptors and preventing virus cell entry. Hence, inhibition of these proteases could be a promising strategy for treatment against SARS-CoV-2. In this review, we discuss the current state of the art of developing inhibitors against the entry proteases furin, the transmembrane serine protease type-II (TMPRSS2), trypsin, and cathepsin L.
Collapse
Affiliation(s)
- Patrick Müller
- Institute for Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, 55128 Mainz, Germany
| | - Hannah Maus
- Institute for Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, 55128 Mainz, Germany
| | - Stefan Josef Hammerschmidt
- Institute for Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, 55128 Mainz, Germany
| | - Philip Knaff
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Tanja Schirmeister
- Institute for Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, 55128 Mainz, Germany
| | - Christian Kersten
- Institute for Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, 55128 Mainz, Germany
| |
Collapse
|
248
|
Wang Q, Li L, Qu T, Li J, Wu L, Li K, Wang Z, Zhu M, Huang B, Wu W, Wu M, Ding R, Zhang Z, Wang Q, Xia X, Li P, Zhang Z, Guo R. High Expression of ACE2 and TMPRSS2 at the Resection Margin Makes Lung Cancer Survivors Susceptible to SARS-CoV-2 With Unfavorable Prognosis. Front Oncol 2021; 11:644575. [PMID: 34094930 PMCID: PMC8176089 DOI: 10.3389/fonc.2021.644575] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) has rapidly spread worldwide. Systematic analysis of lung cancer survivors at molecular and clinical levels is warranted to understand the disease course and clinical characteristics. Methods A single-center, retrospective cohort study was conducted in 65 patients with COVID-19 from Wuhan Huoshenshan Hospital, of which 13 patients were diagnosed with lung cancer. The study was conducted from February 4 to April 11, 2020. Results During the course of treatment, lung cancer survivors infected with severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) had shorter median time from symptom onset to hospitalization (P = 0.016) and longer clinical symptom remission time (P = 0.020) than non-cancer individuals. No differences were observed among indicators such as time from symptom onset to hospitalization and symptom remission time between medium-term and short-term survivors. The expression of ACE2 (P = 0.013) and TMPRSS2 (P <0.001) was elevated in lung cancer survivors as compared with that in non-cancer individuals. Conclusions ACE2 and TMPRSS2 levels were higher at resection margins of lung cancer survivors than those in normal tissues of non-cancerous individuals and may serve as factors responsible for the high susceptibility to COVID-19 among lung cancer survivors. Lung cancer patients diagnosed with COVID-19, including medium-term survivors, have worse outcomes than the general population.
Collapse
Affiliation(s)
- Qianqian Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liangyu Li
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Tianyu Qu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Li
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Lingxiang Wu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Kening Li
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Ziyu Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Mengyan Zhu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Bin Huang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Wei Wu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Min Wu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Rong Ding
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Zhihong Zhang
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qianghu Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Xinyi Xia
- Institute of Laboratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Southern Medical University, Nanjing, China.,Department of Laboratory Medicine & Blood Transfusion, Wuhan Huoshenshan Hospital, Wuhan, China.,Joint Expert Group, Wuhan Huoshenshan Hospital, Wuhan, China
| | - Pengping Li
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Zhi Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Renhua Guo
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
249
|
Halder P, Pal U, Paladhi P, Dutta S, Paul P, Pal S, Das D, Ganguly A, IshitaDutta, SayarneelMandal, Ray A, Ghosh S. Evaluation of potency of the selected bioactive molecules from Indian medicinal plants with M Pro of SARS-CoV-2 through in silico analysis. J Ayurveda Integr Med 2021; 13:100449. [PMID: 34054246 PMCID: PMC8139275 DOI: 10.1016/j.jaim.2021.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 04/12/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background The recent outbreak of novel SARs CoVid-2 across the globe and absence of specific drug against this virus lead the scientific community to look into some alternative indigenous treatments. India as a hub of ayurvedic and medicinal plants can shed light on its treatment using specific active bio-molecules from these plants. Objectives Keeping our herbal resources in mind we were interested to inquire whether some phytochemicals from Indian spices and medicinal plants can be used as alternative therapeutic agents in contrast to synthetic drugs. Materials and methods We used in-silico molecular docking approach to test whether bioactive molecules of herbal origin such as Hyperoside, Nimbaflavone, Ursolic acid, 6-gingerol, 6-shogaol& 6-paradol, Curcumin, Catechins&Epigallocatechin, α-Hederin, Piperine could bind and potentially block theMproenzyme of Sars-CoV-2 virus. Results Ursolic acid showed the highest docking score (-8.7 kcal/mol) followed by Hyperoside (-8.6kcal/mol), α-Hederin (-8.5 kcal/mol) and Nimbaflavone (-8.0kcal/mol). Epigallocatechin, Catechins, and Curcumin also exhibited high binding affinity (Docking score -7.3, -7.1 and -7.1 kcal/mol) with the Mpro. Rest of the tested phytochemicals exhibited moderate binding and inhibitory effects. Conclusion This finding provides a basis for biochemical assay on Sars-CoV-2 virus.
Collapse
Affiliation(s)
- Pinku Halder
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - Upamanyu Pal
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - Pranab Paladhi
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - Saurav Dutta
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - Pallab Paul
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - Samudra Pal
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - Debasmita Das
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - Agnish Ganguly
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - IshitaDutta
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - SayarneelMandal
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| | - Anirban Ray
- Department of Zoology, Bangabasi Morning College (affiliated to University of Calcutta), Kolkata, West Bengal, India, Pincode: 700009
| | - Sujay Ghosh
- Cytogenetics& Genomics Research Unit, Department of Zoology; University of Calcutta, Taraknath-Palit-Siksha-Prangan (Ballygunge Science College Campus), 35 Ballygunge Circular Road, Kolkata, WestBengal, India,Pincode: 700019
| |
Collapse
|
250
|
Rebendenne A, Roy P, Bonaventure B, Chaves Valadão AL, Desmarets L, Rouillé Y, Tauziet M, Arnaud-Arnould M, Giovannini D, Lee Y, DeWeirdt P, Hegde M, Garcia de Gracia F, McKellar J, Wencker M, Dubuisson J, Belouzard S, Moncorgé O, Doench JG, Goujon C. Bidirectional genome-wide CRISPR screens reveal host factors regulating SARS-CoV-2, MERS-CoV and seasonal coronaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34031654 DOI: 10.1101/2021.05.19.444823] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Several genome-wide CRISPR knockout screens have been conducted to identify host factors regulating SARS-CoV-2 replication, but the models used have often relied on overexpression of ACE2 receptor. Additionally, such screens have yet to identify the protease TMPRSS2, known to be important for viral entry at the plasma membrane. Here, we conducted a meta-analysis of these screens and showed a high level of cell-type specificity of the identified hits, arguing for the necessity of additional models to uncover the full landscape of SARS-CoV-2 host factors. We performed genome-wide knockout and activation CRISPR screens in Calu-3 lung epithelial cells, as well as knockout screens in Caco-2 intestinal cells. In addition to identifying ACE2 and TMPRSS2 as top hits, our study reveals a series of so far unidentified and critical host-dependency factors, including the Adaptins AP1G1 and AP1B1 and the flippase ATP8B1. Moreover, new anti-SARS-CoV-2 proteins with potent activity, including several membrane-associated Mucins, IL6R, and CD44 were identified. We further observed that these genes mostly acted at the critical step of viral entry, with the notable exception of ATP8B1, the knockout of which prevented late stages of viral replication. Exploring the pro- and anti-viral breadth of these genes using highly pathogenic MERS-CoV, seasonal HCoV-NL63 and -229E and influenza A orthomyxovirus, we reveal that some genes such as AP1G1 and ATP8B1 are general coronavirus cofactors. In contrast, Mucins recapitulated their known role as a general antiviral defense mechanism. These results demonstrate the value of considering multiple cell models and perturbational modalities for understanding SARS-CoV-2 replication and provide a list of potential new targets for therapeutic interventions.
Collapse
|