1
|
Wang SF, Chen HL, Liu FT. Galectins and Host-Pathogen Interactions: The roles in viral infections. Semin Immunol 2024; 76:101911. [PMID: 39580998 DOI: 10.1016/j.smim.2024.101911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Galectins, a family of carbohydrate-binding proteins, play crucial roles in the host-virus interaction landscape. This review explores the multifaceted contributions of endogenous galectins to various stages of the viral lifecycle, including attachment, replication, assembly, and release of progeny virions. Recent studies have indicated that viral infections can induce the expression and secretion of specific galectins, with elucidated signaling pathways in some cases, enhancing our understanding of their regulatory mechanisms. While many studies have focused on the effects of exogenous recombinant galectins, there is growing interest in the intrinsic functions of endogenous galectins, particularly through genetic alterations in cellular models. This review highlights the need for further research to uncover the complex roles of galectins in modulating viral infections and emphasizes their potential as therapeutic targets in the fight against viral diseases. Understanding these interactions could pave the way for novel strategies to enhance host defense mechanisms and mitigate viral pathogenesis.
Collapse
Affiliation(s)
- Sheng-Fang Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Center for Tropical Medicine and Infectious Disease Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| | - Hung-Lin Chen
- Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Fu-Tong Liu
- Department of Dermatology, Keck School of Medicine of USC, Los Angeles, CA 90033, USA; Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
2
|
Reddy KR, Faridi KF, Aggarwal M, Tirumalai AA, Singh T, Tejtel KS, Williams K, Litwin SE, Dastmalchi LN, White BA, Barnard N, Ornish D, Batts T, Ajene G, Aspry K, Kris Etherton P, Hull SC, Freeman AM. Proposed Mechanisms and Associations of COVID-19 with Cardiometabolic Risk Factors. Am J Lifestyle Med 2024:15598276241269532. [PMID: 39540176 PMCID: PMC11556543 DOI: 10.1177/15598276241269532] [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: 11/16/2024] Open
Abstract
Cardiovascular disease (CVD) and cardiometabolic risk (CMR) are highly prevalent globally. The interplay between CVD/CMR and COVID-19 morbidity and mortality has been intensely studied over the last three years and has yielded some important discoveries and warnings for public health. Despite many advances in cardiovascular medicine, CVD continues to be the global leading cause of death. Much of this disease burden results from high CMR imposed by behaviors centered around poor nutrition related to lifestyle choices and systemic constraints. Increased CVD/CMR contributed to the COVID-19 pandemic's unprecedented wave of disability and death, and the current state of cardiovascular health been equated to a "Population Code Blue." There is an urgent and unmet need to reorient our priorities towards health promotion and disease prevention. This manuscript will review how nutrition and lifestyle affect outcomes in COVID-19 and how some interventions and healthy lifestyle choices can markedly reduce disease burden, morbidity, and mortality.
Collapse
Affiliation(s)
- Koushik R. Reddy
- Division of Cardiology, Department of Medicine, James A. Haley VA Medical Center, University of South Florida, Tampa, FL, USA (KRR)
| | - Kamil F. Faridi
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA (KFF)
| | - Monica Aggarwal
- Division of Cardiology, Department of Medicine, University of Florida, Gainesville, FL, USA (MA)
| | | | - Tamanna Singh
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA (TS)
| | - Kristen S. Tejtel
- Division of Cardiology, Texas Children’s Hospital, Department of Pediatrics, Baylor School of Medicine, Houston, TX, USA (KST)
| | - Kim Williams
- Department of Internal Medicine, University of Louisville, Louisville, KY, USA (KW)
| | - Sheldon E. Litwin
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA (SEL)
| | - Lily Nedda Dastmalchi
- Division of Cardiology, Department of Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA (LND)
| | - Beth Ann White
- Marshall Health, Joan C. Edwards School of Medicine, Huntington, WV, USA (BAW)
| | - Neal Barnard
- Adjunct Faculty, George Washington University School of Medicine & Health Sciences, and Physicians Committee for Responsible Medicine, Washington, DC, USA (NB)
| | - Dean Ornish
- Division of Cardiology and Department of Medicine, Wilford Hall Ambulatory Surgical Center, Uniform Services University, Bethesda, MD, USA
| | - Travis Batts
- Division of Cardiology, Department of Medicine, Wilford Hall Ambulatory Surgical Center, San Antonio, TX, USA (TB)
| | - George Ajene
- Division of Cardiology, Baylor College of Medicine, Houston, TX, USA (GA)
| | - Karen Aspry
- Lifespan Cardiovascular Institute, Alpert Medical School, Brown University, Providence, RI, USA (KA)
| | - Penny Kris Etherton
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA (PKE)
| | - Sarah C. Hull
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA (KFF)
- Program for Biomedical Ethics, Yale School of Medicine, New Haven, CT, USA (SCH)
| | - Andrew M. Freeman
- Division of Cardiology, Department of Medicine, National Jewish Health, Denver, CO, USA (AMF)
| |
Collapse
|
3
|
Jia Q, Yang Y, Yao S, Chen X, Hu Z. Emerging Roles of Galectin-3 in Pulmonary Diseases. Lung 2024; 202:385-403. [PMID: 38850292 DOI: 10.1007/s00408-024-00709-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024]
Abstract
Galectin-3 is a multifunctional protein that is involved in various physiological and pathological events. Emerging evidence suggests that galectin-3 also plays a critical role in the pathogenesis of pulmonary diseases. Galectin-3 can be produced and secreted by various cell types in the lungs, and the overexpression of galectin-3 has been found in acute lung injury/acute respiratory distress syndrome (ALI/ARDS), pulmonary hypertension (PH), pulmonary fibrosis diseases, lung cancer, lung infection, chronic obstructive pulmonary disease (COPD), and asthma. Galectin-3 exerts diverse effects on the inflammatory response, immune cell activation, fibrosis and tissue remodeling, and tumorigenesis in these pulmonary disorders, and genetic and pharmacologic modulation of galectin-3 has therapeutic effects on the treatment of pulmonary illnesses. In this review, we summarize the structure and function of galectin-3 and the underlying mechanisms of galectin-3 in pulmonary disease pathologies; we also discuss preclinical and clinical evidence regarding the therapeutic potential of galectin-3 inhibitors in these pulmonary disorders. Additionally, targeting galectin-3 may be a very promising therapeutic approach for the treatment of pulmonary diseases.
Collapse
Affiliation(s)
- Qi Jia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yiyi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Zhiqiang Hu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
4
|
Saikh KU, Anam K, Sultana H, Ahmed R, Kumar S, Srinivasan S, Ahmed H. Targeting Myeloid Differentiation Primary Response Protein 88 (MyD88) and Galectin-3 to Develop Broad-Spectrum Host-Mediated Therapeutics against SARS-CoV-2. Int J Mol Sci 2024; 25:8421. [PMID: 39125989 PMCID: PMC11313481 DOI: 10.3390/ijms25158421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/16/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Nearly six million people worldwide have died from the coronavirus disease (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although COVID-19 vaccines are largely successful in reducing the severity of the disease and deaths, the decline in vaccine-induced immunity over time and the continuing emergence of new viral variants or mutations underscore the need for an alternative strategy for developing broad-spectrum host-mediated therapeutics against SARS-CoV-2. A key feature of severe COVID-19 is dysregulated innate immune signaling, culminating in a high expression of numerous pro-inflammatory cytokines and chemokines and a lack of antiviral interferons (IFNs), particularly type I (alpha and beta) and type III (lambda). As a natural host defense, the myeloid differentiation primary response protein, MyD88, plays pivotal roles in innate and acquired immune responses via the signal transduction pathways of Toll-like receptors (TLRs), a type of pathogen recognition receptors (PRRs). However, recent studies have highlighted that infection with viruses upregulates MyD88 expression and impairs the host antiviral response by negatively regulating type I IFN. Galectin-3 (Gal3), another key player in viral infections, has been shown to modulate the host immune response by regulating viral entry and activating TLRs, the NLRP3 inflammasome, and NF-κB, resulting in the release of pro-inflammatory cytokines and contributing to the overall inflammatory response, the so-called "cytokine storm". These studies suggest that the specific inhibition of MyD88 and Gal3 could be a promising therapy for COVID-19. This review presents future directions for MyD88- and Gal3-targeted antiviral drug discovery, highlighting the potential to restore host immunity in SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Kamal U. Saikh
- GlycoMantra Inc., bwtech South of the University of Maryland Baltimore County, 1450 South Rolling Road, Baltimore, MD 21227, USA; (K.A.); (H.S.); (R.A.); (S.K.); (S.S.)
| | | | | | | | | | | | - Hafiz Ahmed
- GlycoMantra Inc., bwtech South of the University of Maryland Baltimore County, 1450 South Rolling Road, Baltimore, MD 21227, USA; (K.A.); (H.S.); (R.A.); (S.K.); (S.S.)
| |
Collapse
|
5
|
Kim JS, Ru F, Meeker S, Undem BJ. Direct activation of airway sensory C-fibers by SARS-CoV-2 S1 spike protein. Physiol Rep 2023; 11:e15900. [PMID: 38123162 PMCID: PMC10733116 DOI: 10.14814/phy2.15900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Respiratory viral infection can lead to activation of sensory afferent nerves as indicated by the consequential sore throat, sneezing, coughing, and reflex secretions. In addition to causing troubling symptoms, sensory nerve activation likely accelerates viral spreading. The mechanism how viruses activate sensory nerve terminals during infection is unknown. In this study, we investigate whether coronavirus spike protein activates sensory nerves terminating in the airways. We used isolated vagally-innervated mouse trachea-lung preparation for two-photon microscopy and extracellular electrophysiological recordings. Using two-photon Ca2+ imaging, we evaluated a total number of 786 vagal bronchopulmonary nerves in six experiments. Approximately 49% of the sensory fibers were activated by S1 protein (4 μg/mL intratracheally). Extracellular nerve recording showed the S1 protein evoked action potential discharge in sensory C-fibers; of 39 airway C-fibers (one fiber per mouse), 17 were activated. Additionally, Fura-2 Ca2+ imaging was performed on neurons dissociated from vagal sensory ganglia (n = 254 from 22 mice). The result showed that 63% of neurons responded to S1 protein. SARS-CoV-2 S1 protein can lead to direct activation of sensory C-fiber nerve terminals in the bronchopulmonary tract. Direct activation of C-fibers may contribute to coronavirus symptoms, and amplify viral spreading in a population.
Collapse
Affiliation(s)
- Joyce S. Kim
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fei Ru
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Sonya Meeker
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bradley J. Undem
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
6
|
Das T, Mukhopadhyay C. Comparison and Possible Binding Orientations of SARS-CoV-2 Spike N-Terminal Domain for Gangliosides GM3 and GM1. J Phys Chem B 2023; 127:6940-6948. [PMID: 37523476 DOI: 10.1021/acs.jpcb.3c02286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
SARS-CoV-2 spike glycoprotein is anchored by gangliosides. The sialic acid in the ganglioside headgroup is responsible for virus attachment and entry into host cells. We used coarse-grained (CG) molecular dynamics simulations to expand on our previous study of GM1 interaction with two different orientations of the SARS-CoV-2 S1 subunit N-terminal domain (NTD) and to confirm the role of sialic acid receptors in driving the viral receptor; GM3 was used as another ganglioside on the membrane. Because of the smaller headgroup, sialic acid is crucial in GM3 interactions, whereas GM1 interacts with NTD via both the sialic acid and external galactose. In line with our previous findings for NTD orientations in GM1 binding, we identified two orientations, "compact" and "distributed", comprising sugar receptor-interacting residues in GM3-embedded lipid bilayers. Gangliosides in closer proximity to the compact NTD orientation might cause relatively greater restrictions to penetrate the bilayer. However, the attachment of a distributed NTD orientation with more negative interaction energies appears to facilitate GM1/GM3 to move quickly across the membrane. Our findings likely shed some light on the orientations that the NTD receptor acquires during the early phases of interaction with GM1 and GM3 in a membrane environment.
Collapse
Affiliation(s)
- Tanushree Das
- Department of Chemistry, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Chaitali Mukhopadhyay
- Department of Chemistry, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| |
Collapse
|
7
|
Oliveira ASF, Shoemark DK, Davidson AD, Berger I, Schaffitzel C, Mulholland AJ. SARS-CoV-2 spike variants differ in their allosteric responses to linoleic acid. J Mol Cell Biol 2023; 15:mjad021. [PMID: 36990513 PMCID: PMC10563148 DOI: 10.1093/jmcb/mjad021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 11/07/2022] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
The SARS-CoV-2 spike protein contains a functionally important fatty acid (FA) binding site, which is also found in some other coronaviruses, e.g. SARS-CoV and MERS-CoV. The occupancy of the FA site by linoleic acid (LA) reduces infectivity by 'locking' the spike in a less infectious conformation. Here, we use dynamical-nonequilibrium molecular dynamics (D-NEMD) simulations to compare the allosteric responses of spike variants to LA removal. D-NEMD simulations show that the FA site is coupled to other functional regions of the protein, e.g. the receptor-binding motif (RBM), N-terminal domain (NTD), furin cleavage site, and regions surrounding the fusion peptide. D-NEMD simulations also identify the allosteric networks connecting the FA site to these functional regions. The comparison between the wild-type spike and four variants (Alpha, Delta, Delta plus, and Omicron BA.1) shows that the variants differ significantly in their responses to LA removal. The allosteric connections to the FA site on Alpha are generally similar to those on the wild-type protein, with the exception of the RBM and the S71-R78 region, which show a weaker link to the FA site. In contrast, Omicron is the most different variant, exhibiting significant differences in the RBM, NTD, V622-L629, and furin cleavage site. These differences in the allosteric modulation may be of functional relevance, potentially affecting transmissibility and virulence. Experimental comparison of the effects of LA on SARS-CoV-2 variants, including emerging variants, is warranted.
Collapse
Affiliation(s)
- A Sofia F Oliveira
- School of Chemistry, Centre for Computational Chemistry, University of Bristol, Bristol BS8 1TS, UK
- School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | - Andrew D Davidson
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Imre Berger
- School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- School of Chemistry, Max Planck Bristol Centre for Minimal Biology, Bristol BS8 1TS, UK
| | | | - Adrian J Mulholland
- School of Chemistry, Centre for Computational Chemistry, University of Bristol, Bristol BS8 1TS, UK
| |
Collapse
|
8
|
Pi P, Zeng Z, Zeng L, Han B, Bai X, Xu S. Molecular mechanisms of COVID-19-induced pulmonary fibrosis and epithelial-mesenchymal transition. Front Pharmacol 2023; 14:1218059. [PMID: 37601070 PMCID: PMC10436482 DOI: 10.3389/fphar.2023.1218059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Abstract
As the outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) first broke out in Hubei Province, China, at the end of 2019. It has brought great challenges and harms to global public health. SARS-CoV-2 mainly affects the lungs and is mainly manifested as pulmonary disease. However, one of the biggest crises arises from the emergence of COVID-19-induced fibrosis. At present, there are still many questions about how COVID-19 induced pulmonary fibrosis (PF) occurs and how to treat and regulate its long-term effects. In addition, as an important process of fibrosis, the effect of COVID-19 on epithelial-mesenchymal transition (EMT) may be an important factor driving PF. This review summarizes the main pathogenesis and treatment mechanisms of COVID-19 related to PF. Starting with the basic mechanisms of PF, such as EMT, transforming growth factor-β (TGF-β), fibroblasts and myofibroblasts, inflammation, macrophages, innate lymphoid cells, matrix metalloproteinases and tissue inhibitors of metalloproteinases, hedgehog pathway as well as Notch signaling. Further, we highlight the importance of COVID-19-induced EMT in the process of PF and provide an overview of the related molecular mechanisms, which will facilitate future research to propose new clinical therapeutic solutions for the treatment of COVID-19-induced PF.
Collapse
Affiliation(s)
- Peng Pi
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Zhipeng Zeng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Liqing Zeng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Bing Han
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Xizhe Bai
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Shousheng Xu
- School of Sports Engineering, Beijing Sport University, Beijing, China
| |
Collapse
|
9
|
Sigamani A, Mayo KH, Miller MC, Chen-Walden H, Reddy S, Platt D. An Oral Galectin Inhibitor in COVID-19—A Phase II Randomized Controlled Trial. Vaccines (Basel) 2023; 11:vaccines11040731. [PMID: 37112643 PMCID: PMC10140888 DOI: 10.3390/vaccines11040731] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Background: SARS-CoV-2 vaccines play an important role in reducing disease severity, hospitalization, and death, although they failed to prevent the transmission of SARS-CoV-2 variants. Therefore, an effective inhibitor of galectin-3 (Gal-3) could be used to treat and prevent the transmission of COVID-19. ProLectin-M (PL-M), a Gal-3 antagonist, was shown to interact with Gal-3 and thereby prevent cellular entry of SARS-CoV-2 in previous studies. Aim: The present study aimed to further evaluate the therapeutic effect of PL-M tablets in 34 subjects with COVID-19. Methods: The efficacy of PL-M was evaluated in a randomized, double-blind, placebo-controlled clinical study in patients with mild to moderately severe COVID-19. Primary endpoints included changes in the absolute RT-PCR Ct values of the nucleocapsid and open reading frame (ORF) genes from baseline to days 3 and 7. The incidence of adverse events, changes in blood biochemistry, inflammatory biomarkers, and levels of antibodies against COVID-19 were also evaluated as part of the safety evaluation. Results: PL-M treatment significantly (p = 0.001) increased RT-PCR cycle counts for N and ORF genes on days 3 (Ct values 32.09 ± 2.39 and 30.69 ± 3.38, respectively) and 7 (Ct values 34.91 ± 0.39 and 34.85 ± 0.61, respectively) compared to a placebo treatment. On day 3, 14 subjects in the PL-M group had cycle counts for the N gene above the cut-off value of 29 (target cycle count 29), whereas on day 7, all subjects had cycle counts above the cut-off value. Ct values in placebo subjects were consistently less than 29, and no placebo subjects were RT-PCR-negative until day 7. Most of the symptoms disappeared completely after receiving PL-M treatment for 7 days in more patients compared to the placebo group. Conclusion: PL-M is safe and effective for clinical use in reducing viral loads and promoting rapid viral clearance in COVID-19 patients by inhibiting SARS-CoV-2 entry into cells through the inhibition of Gal-3.
Collapse
Affiliation(s)
- Alben Sigamani
- Carmel Research Consultancy Pvt. Ltd., Bengaluru 560025, Karnataka, India
- Correspondence: ; Tel.: +9188-8443-1444
| | - Kevin H. Mayo
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, Minneapolis, MN 55455, USA
| | - Michelle C. Miller
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, Minneapolis, MN 55455, USA
| | - Hana Chen-Walden
- Pharmalectin India Pvt. Ltd., Rangareddy 500039, Telangana, India
| | - Surendar Reddy
- Department of Pulmonology, ESIC Medical College and Hospital, Sanath Nagar, Hyderabad 500038, Telangana, India
| | - David Platt
- Pharmalectin India Pvt. Ltd., Rangareddy 500039, Telangana, India
| |
Collapse
|
10
|
Mukhina OA, Fomina DS, Parshin VV, Gushchin VA, Dolzhikova IV, Shchetinin AM, Chudakov DM, Alekseeva E, Korostin D, Bazykin GA, Klink G, Logunov DY, Lysenko MA. SARS-CoV-2 evolution in a patient with secondary B-cell immunodeficiency: A clinical case. Hum Vaccin Immunother 2022; 18:2101334. [PMID: 35914217 PMCID: PMC9746374 DOI: 10.1080/21645515.2022.2101334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/27/2022] [Accepted: 07/09/2022] [Indexed: 12/15/2022] Open
Abstract
The article highlights the course of long-term SARS-CoV-2 infection in a patient with a secondary immunodeficiency developed with B-cell-depleting therapy of the underlying disease. Analysis of the intrapatient virus evolution revealed an inpatient S:G75A mutation that alters the 72GTNGTKR78 motif of the S-protein, with a possible role in binding to alternative cellular receptors. Therapy with a ready-made COVID-19-globulin preparation (native human immunoglobulin G (IgG) derived from the plasma of convalescent COVID-19-patients) resulted in rapid improvement of the patient's condition, fast, and stable elimination of the virus, and passive immunization of the patient for at least 30 days. The results suggest the use of products containing neutralizing antibodies opens new prospects for treatment algorithms for patients with persistent coronavirus infection, as well as for passive immunization schemes for patients with a presumably reduced specific response to vaccination.
Collapse
Affiliation(s)
- Olgo A. Mukhina
- Moscow Healthcare Department, State Budgetary Healthcare Institution City Clinical Hospital, Moscow, Russia
| | - Daria S. Fomina
- Moscow Healthcare Department, State Budgetary Healthcare Institution City Clinical Hospital, Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education First Moscow State Medical University named after I.M. Sechenov of the Ministry of Health of the Russian Federation, Sechenov University, Moscow, Russia
| | - Vasiliy V. Parshin
- Moscow Healthcare Department, State Budgetary Healthcare Institution City Clinical Hospital, Moscow, Russia
| | - Vladimir A. Gushchin
- Federal State Budgetary Institution “National Research Center for Epidemiology and Microbiology named after Honorary Academician N.-F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Inna V. Dolzhikova
- Federal State Budgetary Institution “National Research Center for Epidemiology and Microbiology named after Honorary Academician N.-F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexey M. Shchetinin
- Federal State Budgetary Institution “National Research Center for Epidemiology and Microbiology named after Honorary Academician N.-F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Dmitriy M. Chudakov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Evgeniia Alekseeva
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Dmitriy Korostin
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Georgii A. Bazykin
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
- Institute for Information Transmission Problems of the Russian Academy of Sciences (Kharkevich Institute), Moscow, Russia
| | - Galya Klink
- Institute for Information Transmission Problems of the Russian Academy of Sciences (Kharkevich Institute), Moscow, Russia
| | - Denis Yu. Logunov
- Federal State Budgetary Institution “National Research Center for Epidemiology and Microbiology named after Honorary Academician N.-F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Maryana A. Lysenko
- Moscow Healthcare Department, State Budgetary Healthcare Institution City Clinical Hospital, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
11
|
Lam SD, Waman VP, Fraternali F, Orengo C, Lees J. Structural and energetic analyses of SARS-CoV-2 N-terminal domain characterise sugar binding pockets and suggest putative impacts of variants on COVID-19 transmission. Comput Struct Biotechnol J 2022; 20:6302-6316. [PMID: 36408455 PMCID: PMC9639386 DOI: 10.1016/j.csbj.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 is an ongoing pandemic that causes significant health/socioeconomic burden. Variants of concern (VOCs) have emerged affecting transmissibility, disease severity and re-infection risk. Studies suggest that the - N-terminal domain (NTD) of the spike protein may have a role in facilitating virus entry via sialic-acid receptor binding. Furthermore, most VOCs include novel NTD variants. Despite global sequence and structure similarity, most sialic-acid binding pockets in NTD vary across coronaviruses. Our work suggests ongoing evolutionary tuning of the sugar-binding pockets and recent analyses have shown that NTD insertions in VOCs tend to lie close to loops. We extended the structural characterisation of these sugar-binding pockets and explored whether variants could enhance sialic acid-binding. We found that recent NTD insertions in VOCs (i.e., Gamma, Delta and Omicron variants) and emerging variants of interest (VOIs) (i.e., Iota, Lambda and Theta variants) frequently lie close to sugar-binding pockets. For some variants, including the recent Omicron VOC, we find increases in predicted sialic acid-binding energy, compared to the original SARS-CoV-2, which may contribute to increased transmission. These binding observations are supported by molecular dynamics simulations (MD). We examined the similarity of NTD across Betacoronaviruses to determine whether the sugar-binding pockets are sufficiently similar to be exploited in drug design. Whilst most pockets are too structurally variable, we detected a previously unknown highly structurally conserved pocket which can be investigated in pursuit of a generic pan-Betacoronavirus drug. Our structure-based analyses help rationalise the effects of VOCs and provide hypotheses for experiments. Our findings suggest a strong need for experimental monitoring of changes in NTD of VOCs.
Collapse
Affiliation(s)
- Su Datt Lam
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
- Department of Applied Physics, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Vaishali P. Waman
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Franca Fraternali
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Christine Orengo
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Jonathan Lees
- Translational Health Sciences, Bristol Medical University, University of Bristol, Bristol, United Kingdom
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, United Kingdom
| |
Collapse
|
12
|
Abdoli M, Shafaati M, Ghamsari LK, Abdoli A. Intranasal administration of cold-adapted live-attenuated SARS-CoV-2 candidate vaccine confers protection against SARS-CoV-2. Virus Res 2022; 319:198857. [PMID: 35820511 PMCID: PMC9270963 DOI: 10.1016/j.virusres.2022.198857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/15/2022] [Accepted: 07/04/2022] [Indexed: 11/26/2022]
Abstract
With the COVID-19 pandemic globally, the ongoing threat of new challenges of mucosal infections was once again reminded human beings. Hence, access to the next-generation vaccine to elicit mucosal immunity is required to reduce virus shedding. SARS-CoV-2 retains a unique polybasic cleavage motif in its spike protein, recognized by the host furin protease. The proteolytic furin cleavage site at the junction of S1/S2 glycoprotein plays a key role in the pathogenesis of SARS-CoV-2. Here, we examined the protective immunity of a double-deleted PRRA/GTNGTKR motifs cold-adapted live-attenuated candidate vaccines as a called "KaraVac." using a hamster animal model of infected attenuated SARS-CoV-2. The KaraVac vaccinated hamsters were challenged against the wild-type (WT) SARS-CoV-2. No apparent bodyweight loss and histopathological lesions were observed in the hamsters. The establishment of sterilizing immunity was induced via stimulating a robust neutralizing antibody (NAb) response in a hamster model. Consequently, deletions in the spike sequence and inoculation into hamsters provide resistance to the subsequent challenge with WT SARS-CoV-2. We have suggested that deletion of the furin cleavage site and GTNGTKR motifs in the spike sequence attenuates the virus from the parental strain and can be used as a potent immunogen.
Collapse
Affiliation(s)
- Mohsen Abdoli
- Amirabad Virology Laboratory, Vaccine Unit, Tehran 1413693341, Iran
| | - Maryam Shafaati
- Department of Microbiology, Faculty Science, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | | | - Asghar Abdoli
- Amirabad Virology Laboratory, Vaccine Unit, Tehran 1413693341, Iran; Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
13
|
Gulati I, Khan S, Gulati G, Verma SR, Khan M, Ahmad S, Bantun F, Mathkor DM, Haque S. SARS-CoV-2 origins: zoonotic Rhinolophus vs contemporary models. Biotechnol Genet Eng Rev 2022:1-34. [PMID: 36036250 DOI: 10.1080/02648725.2022.2115682] [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: 06/22/2022] [Accepted: 08/03/2022] [Indexed: 11/02/2022]
Abstract
The question of the origin of coronavirus spread like wildfire ever since it wreaked havoc among humankind, and ever since the scientific community has worked tirelessly to trace the history of the virus. In this review, we have tried to compile relevant literature pertaining to the different theories of origin of SARS-CoV-2, hopefully without any bias, and we strongly support the zoonotic origin of the infamous SARS-CoV-2 in bats and its transfer to human beings through the most probable evolutionary hosts, pangolins and minks. We also support the contemporary 'Circulation Model' that simply mirrors the concept of evolution to explain the origin of the virus which, the authors believe, is the most rational school of thought. The most recent variant of SARS-CoV-2, Omicron, has been taken as an example to clarify the concept. We recommend the community to refer to this model for further understanding and delving deep into this mystery of the origin of SARS-CoV-2.
Collapse
Affiliation(s)
- Ishika Gulati
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, Ha'il University, Ha'il, Saudi Arabia
| | - Garima Gulati
- Department of Applied Mechanics, Motilal Nehru National Institute of Technology, Prayagraj, Allahabad, India
| | | | - Mahvish Khan
- Department of Biology, College of science, University of Ha'il, Ha'il, Saudi Arabia
| | - Saheem Ahmad
- Department of clinical laboratory science, College of Applied Medical Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Farkad Bantun
- Department of Microbiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
14
|
Oatis D, Simon-Repolski E, Balta C, Mihu A, Pieretti G, Alfano R, Peluso L, Trotta MC, D’Amico M, Hermenean A. Cellular and Molecular Mechanism of Pulmonary Fibrosis Post-COVID-19: Focus on Galectin-1, -3, -8, -9. Int J Mol Sci 2022; 23:8210. [PMID: 35897786 PMCID: PMC9332679 DOI: 10.3390/ijms23158210] [Citation(s) in RCA: 10] [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: 06/17/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
Pulmonary fibrosis is a consequence of the pathological accumulation of extracellular matrix (ECM), which finally leads to lung scarring. Although the pulmonary fibrogenesis is almost known, the last two years of the COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its post effects added new particularities which need to be explored. Many questions remain about how pulmonary fibrotic changes occur within the lungs of COVID-19 patients, and whether the changes will persist long term or are capable of resolving. This review brings together existing knowledge on both COVID-19 and pulmonary fibrosis, starting with the main key players in promoting pulmonary fibrosis, such as alveolar and endothelial cells, fibroblasts, lipofibroblasts, and macrophages. Further, we provide an overview of the main molecular mechanisms driving the fibrotic process in connection with Galactin-1, -3, -8, and -9, together with the currently approved and newly proposed clinical therapeutic solutions given for the treatment of fibrosis, based on their inhibition. The work underlines the particular pathways and processes that may be implicated in pulmonary fibrosis pathogenesis post-SARS-CoV-2 viral infection. The recent data suggest that galectin-1, -3, -8, and -9 could become valuable biomarkers for the diagnosis and prognosis of lung fibrosis post-COVID-19 and promising molecular targets for the development of new and original therapeutic tools to treat the disease.
Collapse
Affiliation(s)
- Daniela Oatis
- Department of Infectious Disease, Faculty of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
- Doctoral School of Biology, Vasile Goldis Western University of Arad, 310414 Arad, Romania
| | - Erika Simon-Repolski
- Doctoral School of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
- Department of Pneumology, Arad Clinical Emergency Hospital, 310031 Arad, Romania
| | - Cornel Balta
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania;
| | - Alin Mihu
- Department of Microbiology, Faculty of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Gorizio Pieretti
- Department of Plastic Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Roberto Alfano
- Department of Advanced Medical and Surgical Sciences “DAMSS”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Luisa Peluso
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.P.); (M.C.T.); (M.D.)
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.P.); (M.C.T.); (M.D.)
| | - Michele D’Amico
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.P.); (M.C.T.); (M.D.)
| | - Anca Hermenean
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania;
- Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania
| |
Collapse
|
15
|
Buchanan CJ, Gaunt B, Harrison PJ, Yang Y, Liu J, Khan A, Giltrap AM, Le Bas A, Ward PN, Gupta K, Dumoux M, Tan TK, Schimaski L, Daga S, Picchiotti N, Baldassarri M, Benetti E, Fallerini C, Fava F, Giliberti A, Koukos PI, Davy MJ, Lakshminarayanan A, Xue X, Papadakis G, Deimel LP, Casablancas-Antràs V, Claridge TDW, Bonvin AMJJ, Sattentau QJ, Furini S, Gori M, Huo J, Owens RJ, Schaffitzel C, Berger I, Renieri A, Naismith JH, Baldwin AJ, Davis BG. Pathogen-sugar interactions revealed by universal saturation transfer analysis. Science 2022; 377:eabm3125. [PMID: 35737812 DOI: 10.1126/science.abm3125] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Many pathogens exploit host cell-surface glycans. However, precise analyses of glycan ligands binding with heavily modified pathogen proteins can be confounded by overlapping sugar signals and/or compounded with known experimental constraints. Universal saturation transfer analysis (uSTA) builds on existing nuclear magnetic resonance spectroscopy to provide an automated workflow for quantitating protein-ligand interactions. uSTA reveals that early-pandemic, B-origin-lineage severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike trimer binds sialoside sugars in an "end-on" manner. uSTA-guided modeling and a high-resolution cryo-electron microscopy structure implicate the spike N-terminal domain (NTD) and confirm end-on binding. This finding rationalizes the effect of NTD mutations that abolish sugar binding in SARS-CoV-2 variants of concern. Together with genetic variance analyses in early pandemic patient cohorts, this binding implicates a sialylated polylactosamine motif found on tetraantennary N-linked glycoproteins deep in the human lung as potentially relevant to virulence and/or zoonosis.
Collapse
Affiliation(s)
- Charles J Buchanan
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK.,Kavli Institute of Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Ben Gaunt
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK
| | - Peter J Harrison
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, UK.,Diamond Light Source, Harwell Science and Innovation Campus, Oxfordshire, UK
| | - Yun Yang
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, UK
| | - Jiwei Liu
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK
| | - Aziz Khan
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Andrew M Giltrap
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Audrey Le Bas
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, UK
| | - Philip N Ward
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, UK
| | - Kapil Gupta
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol, UK
| | - Maud Dumoux
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK
| | - Tiong Kit Tan
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Lisa Schimaski
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Sergio Daga
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Nicola Picchiotti
- Department of Information Engineering and Mathematics, University of Siena, Siena, Italy.,Department of Mathematics, University of Pavia, Pavia, Italy
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elisa Benetti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Fava
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Annarita Giliberti
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Panagiotis I Koukos
- Bijvoet Centre for Biomolecular Research, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Matthew J Davy
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK
| | - Abirami Lakshminarayanan
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Xiaochao Xue
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK.,Sir William Dunn School of Pathology, Oxford, UK
| | | | | | - Virgínia Casablancas-Antràs
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK.,Kavli Institute of Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | | | - Alexandre M J J Bonvin
- Bijvoet Centre for Biomolecular Research, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | | | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Marco Gori
- Department of Information Engineering and Mathematics, University of Siena, Siena, Italy.,Maasai, I3S CNRS, Université Côte d'Azur, Nice, France
| | - Jiandong Huo
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, UK
| | - Raymond J Owens
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, UK
| | - Christiane Schaffitzel
- Bijvoet Centre for Biomolecular Research, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Imre Berger
- Bijvoet Centre for Biomolecular Research, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy.,Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - James H Naismith
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Headington, Oxford OX3 7BN, UK
| | - Andrew J Baldwin
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK.,Kavli Institute of Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Benjamin G Davis
- Rosalind Franklin Institute, Harwell Science and Innovation Campus, Oxford OX11 0FA, UK.,Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK.,Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
16
|
Schroeder JT, Bieneman AP. The S1 Subunit of the SARS-CoV-2 Spike Protein Activates Human Monocytes to Produce Cytokines Linked to COVID-19: Relevance to Galectin-3. Front Immunol 2022; 13:831763. [PMID: 35392091 PMCID: PMC8982143 DOI: 10.3389/fimmu.2022.831763] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), rapidly evolved into a pandemic –the likes of which has not been experienced in 100 years. While novel vaccines show great efficacy, and therapeutics continue to be developed, the persistence of disease, with the concomitant threat of emergent variants, continues to impose massive health and socioeconomic issues worldwide. Studies show that in susceptible individuals, SARS-CoV-2 infection can rapidly progress toward lung injury and acute respiratory distress syndrome (ARDS), with evidence for an underlying dysregulated innate immune response or cytokine release syndrome (CRS). The mechanisms responsible for this CRS remain poorly understood, yet hyper-inflammatory features were also evident with predecessor viruses within the β-coronaviridae family, namely SARS-CoV-1 and the Middle East Respiratory Syndrome (MERS)-CoV. It is further known that the spike protein (S) of SARS-CoV-2 (as first reported for other β-coronaviruses) possesses a so-called galectin-fold within the N-terminal domain of the S1 subunit (S1-NTD). This fold (or pocket) shows structural homology nearly identical to that of human galectin-3 (Gal-3). In this respect, we have recently shown that Gal-3, when associated with epithelial cells or anchored to a solid phase matrix, facilitates the activation of innate immune cells, including basophils, DC, and monocytes. A synthesis of these findings prompted us to test whether segments of the SARS-CoV-2 spike protein might also activate innate immune cells in a manner similar to that observed in our Gal-3 studies. Indeed, by immobilizing S components onto microtiter wells, we show that only the S1 subunit (with the NTD) activates human monocytes to produce a near identical pattern of cytokines as those reported in COVID-19-related CRS. In contrast, both the S1-CTD/RBD, which binds ACE2, and the S2 subunit (stalk), failed to mediate the same effect. Overall, these findings provide evidence that the SARS-CoV-2 spike protein can activate monocytes for cytokines central to COVID-19, thus providing insight into the innate immune mechanisms underlying the CRS and the potential for therapeutic interventions.
Collapse
Affiliation(s)
- John T Schroeder
- The Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University, Baltimore, MD, United States
| | - Anja P Bieneman
- The Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
17
|
In Silico Analysis of the Multi-Targeted Mode of Action of Ivermectin and Related Compounds. COMPUTATION 2022. [DOI: 10.3390/computation10040051] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Some clinical studies have indicated activity of ivermectin, a macrocyclic lactone, against COVID-19, but a biological mechanism initially proposed for this anti-viral effect is not applicable at physiological concentrations. This in silico investigation explores potential modes of action of ivermectin and 14 related compounds, by which the infectivity and morbidity of the SARS-CoV-2 virus may be limited. Binding affinity computations were performed for these agents on several docking sites each for models of (1) the spike glycoprotein of the virus, (2) the CD147 receptor, which has been identified as a secondary attachment point for the virus, and (3) the alpha-7 nicotinic acetylcholine receptor (α7nAChr), an indicated point of viral penetration of neuronal tissue as well as an activation site for the cholinergic anti-inflammatory pathway controlled by the vagus nerve. Binding affinities were calculated for these multiple docking sites and binding modes of each compound. Our results indicate the high affinity of ivermectin, and even higher affinities for some of the other compounds evaluated, for all three of these molecular targets. These results suggest biological mechanisms by which ivermectin may limit the infectivity and morbidity of the SARS-CoV-2 virus and stimulate an α7nAChr-mediated anti-inflammatory pathway that could limit cytokine production by immune cells.
Collapse
|
18
|
The Impact of COVID-19 on the Food Supply Chain and the Role of E-Commerce for Food Purchasing. SUSTAINABILITY 2022. [DOI: 10.3390/su14053074] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The world has been plagued by an unforeseen threat to public health by the appearance of COVID-19, which has impacted the world’s economy and the worldwide supply chain. The unexpected pandemic (COVID-19) quickly spread across the world, leaving not a single country unaffected by the worldwide pandemic. At the start of COVID-19, several countries adopted social distancing and lockdowns due to the virus. The existing pandemic’s discriminatory aspect has a negative influence on human health and the standard of living, as well as worldwide trade, supply chains, and major economies. COVID-19 has been shown to have an effect throughout the broader food supply chain, one of the largest significant sectors of any country, from the field to the consumers. There are currently substantial concerns regarding food production, manufacturing, delivery, and consumption in light of emerging issues within the food supply chain. Limitations on the mobility of workers, variations in consumer demands, the shutdown of food manufacturing industries, limited food trading regulations, and financial stress throughout the food supply chain are all results of the COVID-19 pandemic. The pandemic (COVID-19) has changed consumer behavior and affected the business and economic sector. The purpose of this study was to explore the application of the Q-technique in the investigation of online consumer and manufacturer behavior in relation to end-user food purchases via digital marketing skills, supply chain possibilities, food purchaser and consumer happiness, and e-commerce infrastructure efficiency under the influence of COVID-19. The results show that in the crisis of supply chain management, clarifying the import food industry e-commerce supply chain crisis items, and determining their priority and strength can help enterprises make emergency decisions regarding supply chain operation; they can also be used as a reference for enterprises to use in responding to the crisis.
Collapse
|
19
|
Sofia F. Oliveira A, Shoemark DK, Avila Ibarra A, Davidson AD, Berger I, Schaffitzel C, Mulholland AJ. The fatty acid site is coupled to functional motifs in the SARS-CoV-2 spike protein and modulates spike allosteric behaviour. Comput Struct Biotechnol J 2021; 20:139-147. [PMID: 34934478 PMCID: PMC8670790 DOI: 10.1016/j.csbj.2021.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
The SARS-CoV-2 spike protein is the first contact point between the SARS-CoV-2 virus and host cells and mediates membrane fusion. Recently, a fatty acid binding site was identified in the spike (Toelzer et al. Science 2020). The presence of linoleic acid at this site modulates binding of the spike to the human ACE2 receptor, stabilizing a locked conformation of the protein. Here, dynamical-nonequilibrium molecular dynamics simulations reveal that this fatty acid site is coupled to functionally relevant regions of the spike, some of them far from the fatty acid binding pocket. Removal of a ligand from the fatty acid binding site significantly affects the dynamics of distant, functionally important regions of the spike, including the receptor-binding motif, furin cleavage site and fusion-peptide-adjacent regions. Simulations of the D614G mutant show differences in behaviour between these clinical variants of the spike: the D614G mutant shows a significantly different conformational response for some structural motifs relevant for binding and fusion. The simulations identify structural networks through which changes at the fatty acid binding site are transmitted within the protein. These communication networks significantly involve positions that are prone to mutation, indicating that observed genetic variation in the spike may alter its response to linoleate binding and associated allosteric communication.
Collapse
Key Words
- ACE2, angiotensin-converting 2 enzyme
- CD, connector domain
- CH, central helix
- FA, fatty acid
- FP, fusion peptide
- FPPR, fusion-peptide proximal region
- HR1, heptad repeat 1
- LA, Linoleic acid
- MD, Molecular dynamics
- MERS, middle east respiratory syndrome
- NTD, N-terminal domain
- RBD, receptor binding domain
- RBM, receptor-binding motif
- RMB, receptor binding motif
- SARS, severe acute respiratory syndrome
- SARS-CoV-2, severe acute respiratory syndrome 2
Collapse
Affiliation(s)
- A. Sofia F. Oliveira
- Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- BrisSynBio, Life Sciences Building, Tyndall Avenue, Bristol BS8 1TQ, UK
| | - Deborah K. Shoemark
- BrisSynBio, Life Sciences Building, Tyndall Avenue, Bristol BS8 1TQ, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Amaurys Avila Ibarra
- Research Software Engineering, Advanced Computing Research Centre, University of Bristol, Bristol BS1 5QD, UK
| | - Andrew D. Davidson
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Imre Berger
- BrisSynBio, Life Sciences Building, Tyndall Avenue, Bristol BS8 1TQ, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Max Planck Bristol Centre for Minimal Biology, Cantock's Close, Bristol BS8 1TS, UK
| | - Christiane Schaffitzel
- BrisSynBio, Life Sciences Building, Tyndall Avenue, Bristol BS8 1TQ, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Adrian J. Mulholland
- Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| |
Collapse
|
20
|
Potential Roles of Modified Pectin Targeting Galectin-3 against Severe Acute Respiratory Syndrome Coronavirus-2. J 2021. [DOI: 10.3390/j4040056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Modified pectin (MP) is a bioactive complex polysaccharide that is broken down into smaller fragments of units and used as an oral dietary supplement for cell proliferation. MP is safe and non-toxic with promising therapeutic properties with regard to targeting galectin-3 (GAL-3) toward the prevention and inhibition of viral infections through the modulation of the immune response and anti-inflammatory cytokine effects. This effect of MP as a GAL-3 antagonism, which has shown benefits in preclinical and clinical models, may be of relevance to the progression of the novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in coronavirus disease 2019 patients. The outbreak of emerging infectious diseases continues to pose a threat to human health. Further to the circulation of multiple variants of SARS-CoV-2, an effective and alternative therapeutic approach to combat it has become pertinent. The use of MP as a GAL-3 inhibitor could serve as an antiviral agent blocking against the SARS-CoV-2-binding spike protein. This review highlights the potential effects of MP in viral infections, its proposed role as a GAL-3 inhibitor, and the associated function concerning a SARS-CoV-2 infection.
Collapse
|
21
|
Du L, Yang Y, Zhang X. Neutralizing antibodies for the prevention and treatment of COVID-19. Cell Mol Immunol 2021; 18:2293-2306. [PMID: 34497376 PMCID: PMC8424621 DOI: 10.1038/s41423-021-00752-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) initiates the infection process by binding to the viral cellular receptor angiotensin-converting enzyme 2 through the receptor-binding domain (RBD) in the S1 subunit of the viral spike (S) protein. This event is followed by virus-cell membrane fusion mediated by the S2 subunit, which allows virus entry into the host cell. Therefore, the SARS-CoV-2 S protein is a key therapeutic target, and prevention and treatment of coronavirus disease 2019 (COVID-19) have focused on the development of neutralizing monoclonal antibodies (nAbs) that target this protein. In this review, we summarize the nAbs targeting SARS-CoV-2 proteins that have been developed to date, with a focus on the N-terminal domain and RBD of the S protein. We also describe the roles that binding affinity, neutralizing activity, and protection provided by these nAbs play in the prevention and treatment of COVID-19 and discuss the potential to improve nAb efficiency against multiple SARS-CoV-2 variants. This review provides important information for the development of effective nAbs with broad-spectrum activity against current and future SARS-CoV-2 strains.
Collapse
Affiliation(s)
- Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA.
| | - Yang Yang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Xiujuan Zhang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| |
Collapse
|
22
|
Aminpour M, Cannariato M, Zucco A, Di Gregorio E, Israel S, Perioli A, Tucci D, Rossi F, Pionato S, Marino S, Deriu MA, Velpula KK, Tuszynski JA. Computational Study of Potential Galectin-3 Inhibitors in the Treatment of COVID-19. Biomedicines 2021; 9:1208. [PMID: 34572394 PMCID: PMC8466820 DOI: 10.3390/biomedicines9091208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
Galectin-3 is a carbohydrate-binding protein and the most studied member of the galectin family. It regulates several functions throughout the body, among which are inflammation and post-injury remodelling. Recent studies have highlighted the similarity between Galectin-3's carbohydrate recognition domain and the so-called "galectin fold" present on the N-terminal domain of the S1 sub-unit of the SARS-CoV-2 spike protein. Sialic acids binding to the N-terminal domain of the Spike protein are known to be crucial for viral entry into humans, and the role of Galectin-3 as a mediator of lung fibrosis has long been the object of study since its levels have been found to be abnormally high in alveolar macrophages following lung injury. In this context, the discovery of a double inhibitor may both prevent viral entry and reduce post-infection pulmonary fibrosis. In this study, we use a database of 56 compounds, among which 37 have known experimental affinity with Galectin-3. We carry out virtual screening of this database with respect to Galectin-3 and Spike protein. Several ligands are found to exhibit promising binding affinity and interaction with the Spike protein's N-terminal domain as well as with Galectin-3. This finding strongly suggests that existing Galectin-3 inhibitors possess dual-binding capabilities to disrupt Spike-ACE2 interactions. Herein we identify the most promising inhibitors of Galectin-3 and Spike proteins, of which five emerge as potential dual effective inhibitors. Our preliminary results warrant further in vitro and in vivo testing of these putative inhibitors against SARS-CoV-2 with the hope of being able to halt the spread of the virus in the future.
Collapse
Affiliation(s)
- Maral Aminpour
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Marco Cannariato
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Angelica Zucco
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Elisabetta Di Gregorio
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Simone Israel
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Annalisa Perioli
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Davide Tucci
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Francesca Rossi
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Sara Pionato
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Silvia Marino
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Marco A. Deriu
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
| | - Kiran K. Velpula
- Department of Cancer Biology and Pharmacology, Pediatrics and Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Jack A. Tuszynski
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; (M.C.); (A.Z.); (E.D.G.); (S.I.); (A.P.); (D.T.); (F.R.); (S.P.); (S.M.); (M.A.D.)
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
23
|
Li X, Zhang L, Chen S, Ji W, Li C, Ren L. Recent progress on the mutations of SARS-CoV-2 spike protein and suggestions for prevention and controlling of the pandemic. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 93:104971. [PMID: 34146731 PMCID: PMC8213438 DOI: 10.1016/j.meegid.2021.104971] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection has caused a global pandemic in the past year, which poses continuing threat to human beings. To date, more than 3561 mutations in the viral spike protein were identified, including 2434 mutations that cause amino acid changes with 343 amino acids located in the viral receptor-binding domain (RBD). Among these mutations, the most representative ones are substitution mutations such as D614G, N501Y, Y453F, N439K/R, P681H, K417N/T, and E484K, and deletion mutations of ΔH69/V70 and Δ242-244, which confer the virus with enhanced infectivity, transmissibility, and resistance to neutralization. In this review, we discussed the recent findings of SARS-CoV-2 for highlighting mutations and variants on virus transmissibility and pathogenicity. Moreover, several suggestions for prevention and controlling the pandemic are also proposed.
Collapse
Affiliation(s)
- Xue Li
- College of Animal Sciences, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, 5333 Xi' An Road, Changchun 130062, China
| | - Liying Zhang
- College of Animal Sciences, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, 5333 Xi' An Road, Changchun 130062, China
| | - Si Chen
- College of Animal Sciences, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, 5333 Xi' An Road, Changchun 130062, China
| | - Weilong Ji
- College of Animal Sciences, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, 5333 Xi' An Road, Changchun 130062, China
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun 130112, China
| | - Linzhu Ren
- College of Animal Sciences, Key Lab for Zoonoses Research, Ministry of Education, Jilin University, 5333 Xi' An Road, Changchun 130062, China.
| |
Collapse
|
24
|
Lechuga GC, Souza-Silva F, Sacramento CQ, Trugilho MRO, Valente RH, Napoleão-Pêgo P, Dias SSG, Fintelman-Rodrigues N, Temerozo JR, Carels N, Alves CR, Pereira MCS, Provance DW, Souza TML, De-Simone SG. SARS-CoV-2 Proteins Bind to Hemoglobin and Its Metabolites. Int J Mol Sci 2021; 22:9035. [PMID: 34445741 PMCID: PMC8396565 DOI: 10.3390/ijms22169035] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/28/2021] [Accepted: 08/10/2021] [Indexed: 01/19/2023] Open
Abstract
(1) Background: coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been linked to hematological dysfunctions, but there are little experimental data that explain this. Spike (S) and Nucleoprotein (N) proteins have been putatively associated with these dysfunctions. In this work, we analyzed the recruitment of hemoglobin (Hb) and other metabolites (hemin and protoporphyrin IX-PpIX) by SARS-Cov2 proteins using different approaches. (2) Methods: shotgun proteomics (LC-MS/MS) after affinity column adsorption identified hemin-binding SARS-CoV-2 proteins. The parallel synthesis of the peptides technique was used to study the interaction of the receptor bind domain (RBD) and N-terminal domain (NTD) of the S protein with Hb and in silico analysis to identify the binding motifs of the N protein. The plaque assay was used to investigate the inhibitory effect of Hb and the metabolites hemin and PpIX on virus adsorption and replication in Vero cells. (3) Results: the proteomic analysis by LC-MS/MS identified the S, N, M, Nsp3, and Nsp7 as putative hemin-binding proteins. Six short sequences in the RBD and 11 in the NTD of the spike were identified by microarray of peptides to interact with Hb and tree motifs in the N protein by in silico analysis to bind with heme. An inhibitory effect in vitro of Hb, hemin, and PpIX at different levels was observed. Strikingly, free Hb at 1mM suppressed viral replication (99%), and its interaction with SARS-CoV-2 was localized into the RBD region of the spike protein. (4) Conclusions: in this study, we identified that (at least) five proteins (S, N, M, Nsp3, and Nsp7) of SARS-CoV-2 recruit Hb/metabolites. The motifs of the RDB of SARS-CoV-2 spike, which binds Hb, and the sites of the heme bind-N protein were disclosed. In addition, these compounds and PpIX block the virus's adsorption and replication. Furthermore, we also identified heme-binding motifs and interaction with hemin in N protein and other structural (S and M) and non-structural (Nsp3 and Nsp7) proteins.
Collapse
Affiliation(s)
- Guilherme C. Lechuga
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
- Laboratory of Celular Ultrastructure, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Franklin Souza-Silva
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
- Biology and Heath Science Faculty, Iguaçu University, Nova Iguaçu 26260-045, RJ, Brazil
| | - Carolina Q. Sacramento
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
- Laboratory of Immunopharmacology, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Monique R. O. Trugilho
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
- Laboratory of Toxinology, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Richard H. Valente
- Laboratory of Toxinology, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Paloma Napoleão-Pêgo
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
| | - Suelen S. G. Dias
- Laboratory of Immunopharmacology, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Natalia Fintelman-Rodrigues
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
- Laboratory of Immunopharmacology, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Jairo R. Temerozo
- Laboratory of Thymus Research, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
- FIOCRUZ, National Institute for Science and Technology on Neuroimmunomodulation (INCT/NIM), Rio de Janeiro 21040-900, RJ, Brazil
| | - Nicolas Carels
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
- Biology and Heath Science Faculty, Iguaçu University, Nova Iguaçu 26260-045, RJ, Brazil
| | - Carlos R. Alves
- Laboratory of Molecular Biology and Endemic Diseases, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Mirian C. S. Pereira
- Laboratory of Celular Ultrastructure, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - David W. Provance
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
| | - Thiago M. L. Souza
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
- Laboratory of Immunopharmacology, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Salvatore G. De-Simone
- FIOCRUZ, Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, RJ, Brazil; (G.C.L.); (F.S.-S.); (C.Q.S.); (M.R.O.T.); (P.N.-P.); (N.F.-R.); (N.C.); (D.W.P.J.); (T.M.L.S.)
- Department of Cellular and Molecular Biology, Biology Institute, Federal Fluminense University, Niterói 24020-141, RJ, Brazil
| |
Collapse
|
25
|
Diagnostic Significance of Serum Galectin-3 in Hospitalized Patients with COVID-19-A Preliminary Study. Biomolecules 2021; 11:biom11081136. [PMID: 34439802 PMCID: PMC8393726 DOI: 10.3390/biom11081136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/15/2021] [Accepted: 07/25/2021] [Indexed: 12/26/2022] Open
Abstract
Severe coronavirus disease 2019 (COVID-19) is associated with hyperinflammation leading to organ injury, including respiratory failure. Galectin-3 was implicated in innate immunological response to infections and in chronic fibrosis. The aim of our preliminary study was the assessment of the diagnostic utility of serum galectin-3 in patients with COVID-19. The prospective observational study included adult patients admitted with active COVID-19 and treated in tertiary hospital between June and July 2020. The diagnosis was confirmed by the quantitative detection of nucleic acid of severe acute respiratory syndrome coronavirus 2 in nasopharyngeal swabs. Galectin-3 was measured by enzyme immunoassay in serum samples obtained during the first five days of hospital stay. We included 70 patients aged 25 to 73 years; 90% had at least one comorbidity. During the hospital stay, 32.9% were diagnosed with COVID-19 pneumonia and 12.9% required treatment in the intensive care unit (ICU). Serum galectin-3 was significantly increased in patients who developed pneumonia, particularly those who required ICU admission. Positive correlations were found between galectin-3 and inflammatory markers (interleukin-6, C-reactive protein, ferritin, pentraxin-3), a marker of endothelial injury (soluble fms-like tyrosine kinase-1), and a range of tissue injury markers. Serum galectin-3 enabled the diagnosis of pneumonia with moderate diagnostic accuracy and the need for ICU treatment with high diagnostic accuracy. Our findings strengthen the hypothesis that galectin-3 may be involved in severe COVID-19. Further studies are planned to confirm the preliminary results and to verify possible associations of galectin-3 with long-term consequences of COVID-19, including pulmonary fibrosis.
Collapse
|
26
|
Di Gaetano S, Capasso D, Delre P, Pirone L, Saviano M, Pedone E, Mangiatordi GF. More Is Always Better Than One: The N-Terminal Domain of the Spike Protein as Another Emerging Target for Hampering the SARS-CoV-2 Attachment to Host Cells. Int J Mol Sci 2021; 22:6462. [PMID: 34208755 PMCID: PMC8235207 DOI: 10.3390/ijms22126462] [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] [Received: 05/14/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 12/31/2022] Open
Abstract
Although the approved vaccines are proving to be of utmost importance in containing the Coronavirus disease 2019 (COVID-19) threat, they will hardly be resolutive as new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, a single-stranded RNA virus) variants might be insensitive to the immune response they induce. In this scenario, developing an effective therapy is still a dire need. Different targets for therapeutic antibodies and diagnostics have been identified, among which the SARS-CoV-2 spike (S) glycoprotein, particularly its receptor-binding domain, has been defined as crucial. In this context, we aim to focus attention also on the role played by the S N-terminal domain (S1-NTD) in the virus attachment, already recognized as a valuable target for neutralizing antibodies, in particular, building on a cavity mapping indicating the presence of two druggable pockets and on the recent literature hypothesizing the presence of a ganglioside-binding domain. In this perspective, we aim at proposing S1-NTD as a putative target for designing small molecules hopefully able to hamper the SARS-CoV-2 attachment to host cells.
Collapse
Affiliation(s)
- Sonia Di Gaetano
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy; (S.D.G.); (L.P.)
- CIRPEB, University of Naples “Federico II”, 80134 Naples, Italy; (D.C.); (M.S.)
| | - Domenica Capasso
- CIRPEB, University of Naples “Federico II”, 80134 Naples, Italy; (D.C.); (M.S.)
- CESTEV, University of Naples “Federico II”, 80145 Naples, Italy
| | - Pietro Delre
- Institute of Crystallography, CNR, 70126 Bari, Italy; (P.D.); (G.F.M.)
- Chemistry Department, University of Bari, 70121 Bari, Italy
| | - Luciano Pirone
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy; (S.D.G.); (L.P.)
| | - Michele Saviano
- CIRPEB, University of Naples “Federico II”, 80134 Naples, Italy; (D.C.); (M.S.)
- Institute of Crystallography, CNR, 70126 Bari, Italy; (P.D.); (G.F.M.)
| | - Emilia Pedone
- Institute of Biostructures and Bioimaging, CNR, 80134 Naples, Italy; (S.D.G.); (L.P.)
- CIRPEB, University of Naples “Federico II”, 80134 Naples, Italy; (D.C.); (M.S.)
| | | |
Collapse
|
27
|
Patro LPP, Sathyaseelan C, Uttamrao PP, Rathinavelan T. The evolving proteome of SARS-CoV-2 predominantly uses mutation combination strategy for survival. Comput Struct Biotechnol J 2021; 19:3864-3875. [PMID: 34109017 PMCID: PMC8178965 DOI: 10.1016/j.csbj.2021.05.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/29/2021] [Accepted: 05/30/2021] [Indexed: 12/31/2022] Open
Abstract
The knowledge about SARS-CoV-2 proteome variations is important to understand its evolutionary tactics and in drug/vaccine design. An extensive analysis of 125,747 whole proteome reveals 7915 recurring mutations (involving 5146 positions) during December2019-November 2020. Among these, 10 and 51 are highly and moderately recurring mutations respectively. Ever since the pandemic outbreak, ∼50% new proteome variants evolve every month, resulting in 5 major clades. Intriguingly, ∼70% of the variants reported in January 2020 are due to the emergence of new mutations, which sharply declines to ∼ 40% in April 2020 and thenceforth, declines steadily till November 2020(∼10%). An exactly opposite trend is seen for variants evolved with cocktail of existing mutations: the lowest in January 2020(∼20%) and the highest in November 2020(80%). This leads to a steady increase in the average number of mutations per sequence. This indicates that the virus has reached the slow pace to accept new mutations. Instead, it uses a mutation combination strategy for survival.
Collapse
Affiliation(s)
| | | | | | - Thenmalarchelvi Rathinavelan
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi Campus, Telangana State 502285, India
| |
Collapse
|
28
|
Jover E, Matilla L, Garaikoetxea M, Fernández-Celis A, Muntendam P, Jaisser F, Rossignol P, López-Andrés N. Beneficial Effects of Mineralocorticoid Receptor Pathway Blockade against Endothelial Inflammation Induced by SARS-CoV-2 Spike Protein. Biomedicines 2021; 9:biomedicines9060639. [PMID: 34204890 PMCID: PMC8228852 DOI: 10.3390/biomedicines9060639] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Vascular endothelial cells activation and dysfunction mediate inflammation and abnormal coagulation in COVID-19 patients. Mineralocorticoid receptor (MR) signaling and its downstream target Galectin-3 (Gal-3) are known to mediate cardiovascular inflammation and might be involved in the pathogenesis of COVID-19 complications. Accordingly, we aimed to investigate the potential beneficial effects of MR antagonism and Gal-3 inhibition on the inflammatory response induced by SARS-CoV-2 Spike protein in human aortic endothelial cells (HAECs). Methods: HAECs were treated with recombinant SARS-COV2 Spike (S) protein. MR antagonists (namely spironolactone and eplerenone) or the Gal-3 inhibitor G3P-01 were supplemented before and after S protein challenge. HAECs supernatants were assessed by ELISA or Western blotting. Results: HAECs treated with recombinant S protein resulted in enhanced secretion of inflammatory molecules (interleukin-6, monocyte chemoattractant protein-1, interleukin-18, interleukin-27, and interferon-γ) as well as in the thrombosis marker plasminogen activator inhibitor (PAI)-1. This was prevented and reversed by both MR antagonists and G3P-01. Conclusions: These findings indicate that MR/Gal-3 pathway blockade could be a promising option to reduce endothelial inflammation in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Eva Jover
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (E.J.); (L.M.); (M.G.); (A.F.-C.)
| | - Lara Matilla
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (E.J.); (L.M.); (M.G.); (A.F.-C.)
| | - Mattie Garaikoetxea
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (E.J.); (L.M.); (M.G.); (A.F.-C.)
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (E.J.); (L.M.); (M.G.); (A.F.-C.)
| | | | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, 75006 Paris, France;
- Centre d’Investigations Cliniques-Plurithématique (INSERM CIC-PT 1433), UMR 1116, CHRU, Université de Lorraine, 54500 Vandoeuvre-Les-Nancy, France;
- French-Clinical Research Infrastructure Network (F-CRIN) Cardiovascular and Renal Clinical Trialists (INI-CRCT), 54500 Nancy, France
| | - Patrick Rossignol
- Centre d’Investigations Cliniques-Plurithématique (INSERM CIC-PT 1433), UMR 1116, CHRU, Université de Lorraine, 54500 Vandoeuvre-Les-Nancy, France;
- French-Clinical Research Infrastructure Network (F-CRIN) Cardiovascular and Renal Clinical Trialists (INI-CRCT), 54500 Nancy, France
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain; (E.J.); (L.M.); (M.G.); (A.F.-C.)
- Correspondence:
| |
Collapse
|
29
|
Schuurs ZP, Hammond E, Elli S, Rudd TR, Mycroft-West CJ, Lima MA, Skidmore MA, Karlsson R, Chen YH, Bagdonaite I, Yang Z, Ahmed YA, Richard DJ, Turnbull J, Ferro V, Coombe DR, Gandhi NS. Evidence of a putative glycosaminoglycan binding site on the glycosylated SARS-CoV-2 spike protein N-terminal domain. Comput Struct Biotechnol J 2021; 19:2806-2818. [PMID: 33968333 PMCID: PMC8093007 DOI: 10.1016/j.csbj.2021.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/01/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022] Open
Abstract
SARS-CoV-2 has rapidly spread throughout the world's population since its initial discovery in 2019. The virus infects cells via a glycosylated spike protein located on its surface. The protein primarily binds to the angiotensin-converting enzyme-2 (ACE2) receptor, using glycosaminoglycans (GAGs) as co-receptors. Here, we performed bioinformatics and molecular dynamics simulations of the spike protein to investigate the existence of additional GAG binding sites on the receptor-binding domain (RBD), separate from previously reported heparin-binding sites. A putative GAG binding site in the N-terminal domain (NTD) of the protein was identified, encompassing residues 245-246. We hypothesized that GAGs of a sufficient length might bridge the gap between this site and the PRRARS furin cleavage site, including the mutation S247R. Docking studies using GlycoTorch Vina and subsequent MD simulations of the spike trimer in the presence of dodecasaccharides of the GAGs heparin and heparan sulfate supported this possibility. The heparan sulfate chain bridged the gap, binding the furin cleavage site and S247R. In contrast, the heparin chain bound the furin cleavage site and surrounding glycosylation structures, but not S247R. These findings identify a site in the spike protein that favors heparan sulfate binding that may be particularly pertinent for a better understanding of the recent UK and South African strains. This will also assist in future targeted therapy programs that could include repurposing clinical heparan sulfate mimetics.
Collapse
Affiliation(s)
- Zachariah P. Schuurs
- QUT, Centre for Genomics and Personalised Health, Cancer and Ageing Research Program, School of Chemistry and Physics, Faculty of Science and Engineering, Institute of Health and Biomedical Innovation, 2 George Street, Brisbane, QLD 4000, Australia
| | - Edward Hammond
- Zucero Therapeutics Ltd, 1 Westlink Court, Brisbane, Queensland, Australia
| | - Stefano Elli
- Istituto di Ricerche Chimiche e Biochimiche “G.Ronzoni”, via Giuseppe Colombo 81, 20133 Milano, Italy
| | - Timothy R. Rudd
- National Institute for Biological Standards and Control, Analytical and Biological Sciences Division, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, UK
| | - Courtney J. Mycroft-West
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Newcastle-Under-Lyme, Staffordshire ST5 5BG, UK
| | - Marcelo A. Lima
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Newcastle-Under-Lyme, Staffordshire ST5 5BG, UK
| | - Mark A. Skidmore
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Newcastle-Under-Lyme, Staffordshire ST5 5BG, UK
| | - Richard Karlsson
- Copenhagen Center for Glycomics, Department of Cellular & Molecular Medicine, University of Copenhagen, Copenhagen N 2200, Denmark
| | - Yen-Hsi Chen
- Copenhagen Center for Glycomics, Department of Cellular & Molecular Medicine, University of Copenhagen, Copenhagen N 2200, Denmark
| | - Ieva Bagdonaite
- Copenhagen Center for Glycomics, Department of Cellular & Molecular Medicine, University of Copenhagen, Copenhagen N 2200, Denmark
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular & Molecular Medicine, University of Copenhagen, Copenhagen N 2200, Denmark
| | - Yassir A. Ahmed
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Derek J. Richard
- QUT, Centre for Genomics and Personalised Health, Cancer & Ageing Research Program, Institute of Health and Biomedical Innovation at the Translational Research Institute (TRI), 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Jeremy Turnbull
- Copenhagen Center for Glycomics, Department of Cellular & Molecular Medicine, University of Copenhagen, Copenhagen N 2200, Denmark
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Deirdre R. Coombe
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Neha S. Gandhi
- QUT, Centre for Genomics and Personalised Health, Cancer and Ageing Research Program, School of Chemistry and Physics, Faculty of Science and Engineering, Institute of Health and Biomedical Innovation, 2 George Street, Brisbane, QLD 4000, Australia
| |
Collapse
|
30
|
Kadam SB, Sukhramani GS, Bishnoi P, Pable AA, Barvkar VT. SARS-CoV-2, the pandemic coronavirus: Molecular and structural insights. J Basic Microbiol 2021; 61:180-202. [PMID: 33460172 PMCID: PMC8013332 DOI: 10.1002/jobm.202000537] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022]
Abstract
The outbreak of a novel coronavirus associated with acute respiratory disease, called COVID-19, marked the introduction of the third spillover of an animal coronavirus (CoV) to humans in the last two decades. The genome analysis with various bioinformatics tools revealed that the causative pathogen (SARS-CoV-2) belongs to the subgenus Sarbecovirus of the genus Betacoronavirus, with highly similar genome as bat coronavirus and receptor-binding domain (RBD) of spike glycoprotein as Malayan pangolin coronavirus. Based on its genetic proximity, SARS-CoV-2 is likely to have originated from bat-derived CoV and transmitted to humans via an unknown intermediate mammalian host, probably Malayan pangolin. Further, spike protein S1/S2 cleavage site of SARS-CoV-2 has acquired polybasic furin cleavage site which is absent in bat and pangolin suggesting natural selection either in an animal host before zoonotic transfer or in humans following zoonotic transfer. In the current review, we recapitulate a preliminary opinion about the disease, origin and life cycle of SARS-CoV-2, roles of virus proteins in pathogenesis, commonalities, and differences between different corona viruses. Moreover, the crystal structures of SARS-CoV-2 proteins with unique characteristics differentiating it from other CoVs are discussed. Our review also provides comprehensive information on the molecular aspects of SARS-CoV-2 including secondary structures in the genome and protein-protein interactions which can be useful to understand the aggressive spread of the SARS-CoV-2. The mutations and the haplotypes reported in the SARS-CoV-2 genome are summarized to understand the virus evolution.
Collapse
Affiliation(s)
| | | | | | - Anupama A. Pable
- Department of MicrobiologySavitribai Phule Pune UniversityPuneIndia
| | | |
Collapse
|
31
|
Lobo-Galo N, Gálvez-Ruíz JC, Balderrama-Carmona AP, Silva-Beltrán NP, Ruiz-Bustos E. Recent biotechnological advances as potential intervention strategies against COVID-19. 3 Biotech 2021; 11:41. [PMID: 33457170 PMCID: PMC7796695 DOI: 10.1007/s13205-020-02619-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
The emerging SARS-CoV-2 viral disease (COVID-19) has caused a global health alert due to its high rate of infection and mortality in individuals with chronic cardiovascular comorbidities, in addition to generating complex clinical conditions. This has forced the scientific community to explore different strategies that allow combating this viral infection as well as treating life-threatening systemic effect of the infection on the individual. In this work, we have reviewed the most recent scientific evidence to provide a comprehensive panorama regarding the biotechnological strategies that have been proposed to combat this new viral infection. We have focused our analysis on vaccine production, nanotechnology applications, repurposing of know drugs for unrelated pathologies, and the search for bioactive molecules obtained from natural products. The goals include safely use as potential prophylactic or therapeutic treatments, based on in silico and in vivo studies, including clinical trials around the world for the correct and timely diagnosis of the infection. This review aims to highlight the development of new ideas that can decrease the time lines for research output and improve research quality while at the same time, keeping in mind the efficacy and safety aspects of these potential biotechnological strategies.
Collapse
Affiliation(s)
- Naun Lobo-Galo
- Departamento de Ciencias Químico Biológicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua Mexico
| | - Juan-Carlos Gálvez-Ruíz
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora, Hermosillo, Sonora Mexico
| | - Ana P. Balderrama-Carmona
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Unidad Regional Sur, Universidad de Sonora, Navojoa, Sonora Mexico
| | - Norma P. Silva-Beltrán
- Departamento de Ciencias de la Salud, campus Cajeme, Universidad de Sonora, Ciudad Obregón, Sonora Mexico
| | - Eduardo Ruiz-Bustos
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora, Hermosillo, Sonora Mexico
| |
Collapse
|
32
|
Seyran M, Takayama K, Uversky VN, Lundstrom K, Palù G, Sherchan SP, Attrish D, Rezaei N, Aljabali AAA, Ghosh S, Pizzol D, Chauhan G, Adadi P, Mohamed Abd El-Aziz T, Soares AG, Kandimalla R, Tambuwala M, Hassan SS, Azad GK, Pal Choudhury P, Baetas-da-Cruz W, Serrano-Aroca Á, Brufsky AM, Uhal BD. The structural basis of accelerated host cell entry by SARS-CoV-2†. FEBS J 2020; 288:5010-5020. [PMID: 33264497 PMCID: PMC7753708 DOI: 10.1111/febs.15651] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) is the causative agent of the pandemic coronavirus disease 2019 (COVID‐19) that exhibits an overwhelming contagious capacity over other human coronaviruses (HCoVs). This structural snapshot describes the structural bases underlying the pandemic capacity of SARS‐CoV‐2 and explains its fast motion over respiratory epithelia that allow its rapid cellular entry. Based on notable viral spike (S) protein features, we propose that the flat sialic acid‐binding domain at the N‐terminal domain (NTD) of the S1 subunit leads to more effective first contact and interaction with the sialic acid layer over the epithelium, and this, in turn, allows faster viral ‘surfing’ of the epithelium and receptor scanning by SARS‐CoV‐2. Angiotensin‐converting enzyme 2 (ACE‐2) protein on the epithelial surface is the primary entry receptor for SARS‐CoV‐2, and protein–protein interaction assays demonstrate high‐affinity binding of the spike protein (S protein) to ACE‐2. To date, no high‐frequency mutations were detected at the C‐terminal domain of the S1 subunit in the S protein, where the receptor‐binding domain (RBD) is located. Tight binding to ACE‐2 by a conserved viral RBD suggests the ACE2‐RBD interaction is likely optimal. Moreover, the viral S subunit contains a cleavage site for furin and other proteases, which accelerates cell entry by SARS‐CoV‐2. The model proposed here describes a structural basis for the accelerated host cell entry by SARS‐CoV‐2 relative to other HCoVs and also discusses emerging hypotheses that are likely to contribute to the development of antiviral strategies to combat the pandemic capacity of SARS‐CoV‐2.
Collapse
Affiliation(s)
- Murat Seyran
- Doctoral Studies in Natural and Technical Sciences (SPL 44), University of Vienna, Austria.,Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Vienna, Austria
| | - Kazuo Takayama
- Center for iPS Cell Research and Application, Kyoto University, Japan
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Italy
| | - Samendra P Sherchan
- Department of Environmental Health Sciences, Tulane University, New Orleans, LA, USA
| | - Diksha Attrish
- Dr. B R Ambedkar Center for Biomedical Research (ACBR), University of Delhi (North Camps), Delhi, India
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Shinjini Ghosh
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, Kolkata, India
| | - Damiano Pizzol
- Italian Agency for Development Cooperation - Khartoum, Al Amarat, Sudan
| | - Gaurav Chauhan
- School of Engineering and Sciences, Tecnologico de Monterrey, Mexico
| | - Parise Adadi
- Department of Food Science, University of Otago, Dunedin, New Zealand
| | - Tarek Mohamed Abd El-Aziz
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA.,Zoology Department, Faculty of Science, Minia University, El-Minia, Egypt
| | - Antonio G Soares
- Zoology Department, Faculty of Science, Minia University, El-Minia, Egypt
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, India.,Department of Biochemistry, Kakatiya Medical College, Warangal, India
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, UK
| | - Sk Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Paschim Medinipur, India
| | | | | | - Wagner Baetas-da-Cruz
- Translational Laboratory in Molecular Physiology, Centre for Experimental Surgery, College of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Adam M Brufsky
- Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bruce D Uhal
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
33
|
Caniglia JL, Asuthkar S, Tsung AJ, Guda MR, Velpula KK. Immunopathology of galectin-3: an increasingly promising target in COVID-19. F1000Res 2020; 9:1078. [PMID: 33082935 PMCID: PMC7536583 DOI: 10.12688/f1000research.25979.2] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 01/18/2023] Open
Abstract
The pandemic brought on by the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has become a global health crisis, with over 22 million confirmed cases and 777,000 fatalities due to coronavirus disease 2019 (COVID-19) reported worldwide. The major cause of fatality in infected patients, now referred to as the "Cytokine Storm Syndrome" (CSS), is a direct result of aberrant immune activation following SARS-CoV2 infection and results in excess release of inflammatory cytokines, such as interleukin (IL)-1, tumor necrosis factor α (TNF-α), and IL-6, by macrophages, monocytes, and dendritic cells. Single cell analysis has also shown significantly elevated levels of galectin 3 (Gal-3) in macrophages, monocytes, and dendritic cells in patients with severe COVID-19 as compared to mild disease. Inhibition of Gal-3 reduces the release of IL-1, IL-6, and TNF-α from macrophages in vitro, and as such may hold promise in reducing the incidence of CSS. In addition, Gal-3 inhibition shows promise in reducing transforming growth factor ß (TGF-ß) mediated pulmonary fibrosis, likely to be a major consequence in survivors of severe COVID-19. Finally, a key domain in the spike protein of SARS-CoV2 has been shown to bind N-acetylneuraminic acid (Neu5Ac), a process that may be essential to cell entry by the virus. This Neu5Ac-binding domain shares striking morphological, sequence, and functional similarities with human Gal-3. Here we provide an updated review of the literature linking Gal-3 to COVID-19 pathogenesis. Dually targeting galectins and the Neu5Ac-binding domain of SARS-CoV2 shows tentative promise in several stages of the disease: preventing viral entry, modulating the host immune response, and reducing the post-infectious incidence of pulmonary fibrosis.
Collapse
Affiliation(s)
- John L. Caniglia
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Swapna Asuthkar
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Andrew J. Tsung
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Illinois Neurological Institute, Peoria, IL, USA
| | - Maheedhara R. Guda
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Kiran K. Velpula
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| |
Collapse
|
34
|
Caniglia JL, Asuthkar S, Tsung AJ, Guda MR, Velpula KK. Immunopathology of galectin-3: an increasingly promising target in COVID-19. F1000Res 2020; 9:1078. [PMID: 33082935 PMCID: PMC7536583 DOI: 10.12688/f1000research.25979.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 01/11/2024] Open
Abstract
The pandemic brought on by the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has become a global health crisis, with over 22 million confirmed cases and 777,000 fatalities due to coronavirus disease 2019 (COVID-19) reported worldwide. The major cause of fatality in infected patients, now referred to as the "Cytokine Storm Syndrome" (CSS), is a direct result of aberrant immune activation following SARS-CoV2 infection and results in excess release of inflammatory cytokines, such as interleukin (IL)-1, tumor necrosis factor α (TNF-α), and IL-6, by macrophages, monocytes, and dendritic cells. Single cell analysis has also shown significantly elevated levels of galectin 3 (Gal-3) in macrophages, monocytes, and dendritic cells in patients with severe COVID-19 as compared to mild disease. Inhibition of Gal-3 reduces the release of IL-1, IL-6, and TNF-α from macrophages in vitro, and as such may hold promise in reducing the incidence of CSS. In addition, Gal-3 inhibition shows promise in reducing transforming growth factor ß (TGF-ß) mediated pulmonary fibrosis, likely to be a major consequence in survivors of severe COVID-19. Finally, a key domain in the spike protein of SARS-CoV2 has been shown to bind N-acetylneuraminic acid (Neu5Ac), a process that may be essential to cell entry by the virus. This Neu5Ac-binding domain shares striking morphological, sequence, and functional similarities with human Gal-3. Here we provide an updated review of the literature linking Gal-3 to COVID-19 pathogenesis. Dually targeting galectins and the Neu5Ac-binding domain of SARS-CoV2 shows tentative promise in several stages of the disease: preventing viral entry, modulating the host immune response, and reducing the post-infectious incidence of pulmonary fibrosis.
Collapse
Affiliation(s)
- John L. Caniglia
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Swapna Asuthkar
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Andrew J. Tsung
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Illinois Neurological Institute, Peoria, IL, USA
| | - Maheedhara R. Guda
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Kiran K. Velpula
- Departments of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
- Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| |
Collapse
|
35
|
Awasthi M, Gulati S, Sarkar DP, Tiwari S, Kateriya S, Ranjan P, Verma SK. The Sialoside-Binding Pocket of SARS-CoV-2 Spike Glycoprotein Structurally Resembles MERS-CoV. Viruses 2020; 12:E909. [PMID: 32825063 PMCID: PMC7551769 DOI: 10.3390/v12090909] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
COVID-19 novel coronavirus (CoV) disease caused by severe acquired respiratory syndrome (SARS)-CoV-2 manifests severe lethal respiratory illness in humans and has recently developed into a worldwide pandemic. The lack of effective treatment strategy and vaccines against the SARS-CoV-2 poses a threat to human health. An extremely high infection rate and multi-organ secondary infection within a short period of time makes this virus more deadly and challenging for therapeutic interventions. Despite high sequence similarity and utilization of common host-cell receptor, human angiotensin-converting enzyme-2 (ACE2) for virus entry, SARS-CoV-2 is much more infectious than SARS-CoV. Structure-based sequence comparison of the N-terminal domain (NTD) of the spike protein of Middle East respiratory syndrome (MERS)-CoV, SARS-CoV, and SARS-CoV-2 illustrate three divergent loop regions in SARS-CoV-2, which is reminiscent of MERS-CoV sialoside binding pockets. Comparative binding analysis with host sialosides revealed conformational flexibility of SARS-CoV-2 divergent loop regions to accommodate diverse glycan-rich sialosides. These key differences with SARS-CoV and similarity with MERS-CoV suggest an evolutionary adaptation of SARS-CoV-2 spike glycoprotein reciprocal interaction with host surface sialosides to infect host cells with wide tissue tropism.
Collapse
Affiliation(s)
- Mayanka Awasthi
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA;
| | | | - Debi P. Sarkar
- Department of Biochemistry, University of Delhi South Campus, New Delhi 110021, India;
| | - Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India;
| | - Suneel Kateriya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Peeyush Ranjan
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA;
| | - Santosh Kumar Verma
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India;
| |
Collapse
|