101
|
Identification of novel regulatory pathways across normal human bronchial epithelial cell lines (NHBEs) and peripheral blood mononuclear cell lines (PBMCs) in COVID-19 patients using transcriptome analysis. INFORMATICS IN MEDICINE UNLOCKED 2022; 31:100979. [PMID: 35669390 PMCID: PMC9159965 DOI: 10.1016/j.imu.2022.100979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
The SARS-CoV-2 is one of the most infectious and deadly coronaviruses, which has gripped the world, causing the COVID-19 pandemic. Despite the numerous studies being conducted on this virus, many uncertainties are with the disease. This is exacerbated by the speedy mutations acquired by the viral strain, which enables the disease to present itself differently in different people, introducing new factors of uncertainty. This study aims at the identification of regulatory pathways across two cell lines, namely, the peripheral blood mononuclear cell line (PBMC) and the normal human bronchial epithelial (NHBE) cell line. Both the above-mentioned cell lines were considered because they support viral replication. Furthermore, the NHBE cell line captures vital changes in the lungs, which are the main organs affected by the COVID-19 patients, and the PBMC cell line is closely linked to the body's immune system. RNA-Seq analysis, differential gene expression and gene set enrichment analysis for pathway identification were followed. Pathway analysis throws light upon the various systems affected in the body due to the COVID-19. Gene regulatory networks associated with the significant pathways were also designed. These networks aid in identifying various gene targets, along with their interactions. Studying the functionality of the pathways and the gene interactions associated with them, aided by long COVID studies, will provide immense clarity about the current COVID-19 scenario. In the long term, this will help in the design of therapeutic approaches against the SARS-CoV-2 and can also contribute to drug repurposing studies. Ultimately, this study identifies and analyses the relationship of various undiscovered or lesser explored pathways in the human body to the SARS-CoV-2 and establish a clearer picture of the association to help streamline further studies and approaches.
Collapse
|
102
|
Smoak P, Burke SJ, Martin TM, Batdorf HM, Floyd ZE, Collier JJ. Artemisia dracunculus L. Ethanolic Extract and an Isolated Component, DMC2, Ameliorate Inflammatory Signaling in Pancreatic β-Cells via Inhibition of p38 MAPK. Biomolecules 2022; 12:biom12050708. [PMID: 35625635 PMCID: PMC9139089 DOI: 10.3390/biom12050708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Non-resolving pancreatic islet inflammation is widely viewed as a contributor to decreases in β-cell mass and function that occur in both Type 1 and Type 2 diabetes. Therefore, strategies aimed at reducing or eliminating pathological inflammation would be useful to protect islet β-cells. Herein, we described the use of 2′,4′-dihydroxy-4-methoxydihydrochalcone (DMC2), a bioactive molecule isolated from an ethanolic extract of Artemisia dracunculus L., as a novel anti-inflammatory agent. The ethanolic extract, termed PMI 5011, reduced IL-1β-mediated NF-κB activity. DMC2 retained this ability, indicating this compound as the likely source of anti-inflammatory activity within the overall PMI 5011 extract. We further examined NF-κB activity using promoter-luciferase reporter constructs, Western blots, mRNA abundance, and protein secretion. Specifically, we found that PMI 5011 and DMC2 each reduced the ability of IL-1β to promote increases in the expression of the Ccl2 and Ccl20 genes. These genes encode proteins that promote immune cell recruitment and are secreted by β-cells in response to IL-1β. Phosphorylation of IκBα and the p65 subunit of NF-κB were not reduced by either PMI 5011 or DMC2; however, phosphorylation of p38 MAPK was blunted in the presence of DMC2. Finally, we observed that while PMI 5011 impaired glucose-stimulated insulin secretion, insulin output was preserved in the presence of DMC2. In conclusion, PMI 5011 and DMC2 reduced inflammation, but only DMC2 did so with the preservation of glucose-stimulated insulin secretion.
Collapse
Affiliation(s)
- Peter Smoak
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (P.S.); (T.M.M.); (H.M.B.)
| | - Susan J. Burke
- Laboratory of Immunogenetics, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA;
| | - Thomas M. Martin
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (P.S.); (T.M.M.); (H.M.B.)
| | - Heidi M. Batdorf
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (P.S.); (T.M.M.); (H.M.B.)
| | - Z. Elizabeth Floyd
- Laboratory of Ubitquitin Biology, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA;
| | - J. Jason Collier
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; (P.S.); (T.M.M.); (H.M.B.)
- Correspondence:
| |
Collapse
|
103
|
Balakrishna Pillai A, JeanPierre AR, Mariappan V, Ranganadin P, S R R. Neutralizing the free radicals could alleviate the disease severity following an infection by positive strand RNA viruses. Cell Stress Chaperones 2022; 27:189-195. [PMID: 35366756 PMCID: PMC8976658 DOI: 10.1007/s12192-022-01269-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
Free radical release due to oxidative stress is gaining importance in the field of viral pathogenesis. Recent studies suggest the involvement of oxidative stress and ROS levels in regulating disease virulence during RNA virus infection. Most of the RNA virus infections lead to vascular dysfunction and disease severity. However, the biology of free radicals in maintaining vascular endothelium integrity is not completely understood. In the present review, we discuss some of the common features in positive-strand RNA virus infections such as dengue and SARS-CoV-2 and suggest that anti-oxidant therapy could pave the way to develop therapeutic strategies in combating emerging and re-emerging RNA viruses.
Collapse
Affiliation(s)
- Agieshkumar Balakrishna Pillai
- Central Inter-Disciplinary Research Facility (CIDRF), School of Biological Sciences, MGM Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed To Be University), Puducherry, 607 402, India.
| | - Aashika Raagavi JeanPierre
- Central Inter-Disciplinary Research Facility (CIDRF), School of Biological Sciences, MGM Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed To Be University), Puducherry, 607 402, India
| | - Vignesh Mariappan
- Central Inter-Disciplinary Research Facility (CIDRF), School of Biological Sciences, MGM Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed To Be University), Puducherry, 607 402, India
| | - Pajanivel Ranganadin
- Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed To Be University), Puducherry, 607 402, India
| | - Rao S R
- Research, Innovation & Development, Sri Balaji Vidyapeeth (Deemed To Be University), Puducherry, 607 402, India
| |
Collapse
|
104
|
Al-Kuraishy HM, Al-Gareeb AI, Butnariu M, Batiha GES. The crucial role of prolactin-lactogenic hormone in Covid-19. Mol Cell Biochem 2022; 477:1381-1392. [PMID: 35147901 PMCID: PMC8831165 DOI: 10.1007/s11010-022-04381-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022]
Abstract
Prolactin (PRL) is a peptide hormone secreted from anterior pituitary involved in milk production in the females and regulation of sex drive in both sexes. PRL has pro-inflammatory and anti-inflammatory functions. High PRL serum level or hyperprolactinemia is associated with different viral infections. In coronavirus disease 2019 (Covid-19), which caused by positive-sense single-strand RNA virus known as severe acute respiratory distress syndrome coronavirus type 2 (SARS-CoV-2), PRL serum level is increased. PRL in Covid-19 may exacerbate the underlying inflammatory status by induction release of pro-inflammatory cytokines. However, PRL through its anti-inflammatory effects may reduce the hyperinflammatory status in Covid-19. The underlying mechanism of increasing PRL in Covid-19 is poorly understood. Therefore, in this review we try to find the potential anti-inflammatory or pro-inflammatory role of PRL in Covid-19. As well, this review was aimed to discuss the underlying causes and mechanisms for Covid-19-induced hyperprolactinemia.
Collapse
Affiliation(s)
| | - Ali I. Al-Gareeb
- College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Monica Butnariu
- Chemistry & Biochemistry Discipline, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” From Timisoara, Calea Aradului 119, 300645 Timis, Romania
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511 Egypt
| |
Collapse
|
105
|
Braga CL, Acquarone M, Arona VDC, Osório BS, Barreto TG, Kian RM, Pereira JPAL, Silva MDMCD, Silva BA, de Oliveira GMM, Macedo Rocco PR, Silva PL, Alencar AKN. Can Epigenetics Help Solve the Puzzle Between Concomitant Cardiovascular Injury and Severity of Coronavirus Disease 2019? J Cardiovasc Pharmacol 2022; 79:431-443. [PMID: 34935698 DOI: 10.1097/fjc.0000000000001201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/29/2021] [Indexed: 01/08/2023]
Abstract
ABSTRACT The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by SARS-CoV-2 has significant implications in patients with concomitant cardiovascular disease (CVD) because they are the population at the greatest risk of death. The treatment of such patients and complications may represent a new challenge for the fields of cardiology and pharmacology. Thus, understanding the involvement of this viral infection in CVD might help to reduce the aggressiveness of SARS-CoV-2 in causing multiorgan infection and damage. SARS-CoV-2 disturbs the host epigenome and several epigenetic processes involved in the pathophysiology of COVID-19 that can directly affect the function and structure of the cardiovascular system (CVS). Hence, it would be relevant to identify epigenetic alterations that directly impact CVS physiology after SARS-CoV-2 infection. This could contribute to the view of this virus-induced CVS injury and direct forthcoming tackles for COVID-19 treatment to reduce mortality in patients with CVD. Targeting epigenetic marks could offer strong evidence for the development of novel antiviral therapies, especially in the context of COVID-19-related CVS damage. In this review, we address some of the main signaling pathways that are currently known as being involved in COVID-19 pathophysiology and the importance of this glint on epigenetics and some of its modifiers (epidrugs) to control the unregulated epitope activity in the context of SARS-CoV-2 infection, COVID-19, and underlying CVD.
Collapse
Affiliation(s)
- Cássia L Braga
- Laboratório de Investigação Pulmonar, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Acquarone
- Faculdade de Medicina de Petrópolis, School Clinic, Petrópolis, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor da C Arona
- Faculdade de Medicina de Petrópolis, School Clinic, Petrópolis, Brazil
| | - Brenno S Osório
- Faculdade de Medicina de Petrópolis, School Clinic, Petrópolis, Brazil
| | - Thiago G Barreto
- Faculdade de Medicina de Petrópolis, School Clinic, Petrópolis, Brazil
| | - Ruan M Kian
- Faculdade de Medicina de Petrópolis, School Clinic, Petrópolis, Brazil
| | | | - Marina de Moraes C da Silva
- Serviço de Radiologia do Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bagnólia A Silva
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Departamento de Ciências Farmacêuticas, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Gláucia Maria M de Oliveira
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; and
| | - Patricia Rieken Macedo Rocco
- Laboratório de Investigação Pulmonar, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratório de Investigação Pulmonar, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Allan K N Alencar
- Laboratório de Investigação Pulmonar, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Faculdade de Medicina de Petrópolis, School Clinic, Petrópolis, Brazil
- Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
106
|
Wang J, Ge W, Peng X, Yuan L, He S, Fu X. Investigating the active compounds and mechanism of HuaShi XuanFei formula for prevention and treatment of COVID-19 based on network pharmacology and molecular docking analysis. Mol Divers 2022; 26:1175-1190. [PMID: 34105049 PMCID: PMC8187140 DOI: 10.1007/s11030-021-10244-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/03/2021] [Indexed: 01/13/2023]
Abstract
Traditional Chinese medicine (TCM) has exerted positive effects in controlling the COVID-19 pandemic. HuaShi XuanFei Formula (HSXFF) was developed to treat patients with mild and general COVID-19 in Zhejiang Province, China. The present study seeks to explore its potentially active compounds and pharmacological mechanisms against COVID-19 based on network pharmacology, molecular docking, and molecular dynamics (MD) simulation. All components of HSXFF were harvested from the pharmacology database of the TCMSP system. COVID-19-related targets were retrieved from using OMIM and GeneCards databases. The herb-compound-targets network was constructed by Cytoscape. The target protein-protein interaction (PPI) network, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed to discover the potential key target genes and mechanism. The main active compounds of HSXFF were docked with 3C-like (3CL) protease hydrolase and angiotensin-converting enzyme 2 (ACE2). The MD simulation confirmed the binding stability of docking results. The herbs-targets network mainly contained 52 compounds and 70 corresponding targets, including key targets such as RELA, TNF, TP53, IL6, MAPK1, CXCL8, IL-1β, and MAPK14. The GO and KEGG indicated that HSXFF may be mainly acting on the IL-17 signaling pathway, TNF signaling pathway, NF-κB signaling pathway, etc. The molecular docking results indicated that isovitexin and procyanidin B1 showed the highest affinity with 3CL and ACE2, respectively, which were confirmed by MD simulation. These findings suggested HSXFF exerted therapeutic effects involving "multi-compounds and multi-targets." It might be working through directly inhibiting the virus, improving immune function, and reducing the inflammatory in response to anti-COVID-19. In summary, the present study would provide a valuable direction for further research of HSXFF.
Collapse
Affiliation(s)
- Juan Wang
- Zhejiang Pharmaceutical College, Zhejiang Province, Ningbo, 315100, People's Republic of China
| | - Wen Ge
- Zhejiang Pharmaceutical College, Zhejiang Province, Ningbo, 315100, People's Republic of China
| | - Xin Peng
- Ningbo Research Institute of Zhejiang University, Zhejiang Province, Ningbo, 315100, People's Republic of China.
| | - Lixia Yuan
- Zhejiang Pharmaceutical College, Zhejiang Province, Ningbo, 315100, People's Republic of China
| | - Shuaibing He
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou Central Hospital, Huzhou, People's Republic of China
| | - Xuyan Fu
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou Central Hospital, Huzhou, People's Republic of China
| |
Collapse
|
107
|
Kim J, Im YN, Chung Y, Youm J, Im SY, Han MK, Lee HK. Glutamine deficiency shifts the asthmatic state toward neutrophilic airway inflammation. Allergy 2022; 77:1180-1191. [PMID: 34601745 PMCID: PMC9293426 DOI: 10.1111/all.15121] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/04/2022]
Abstract
Background The administration of L‐glutamine (Gln) suppresses allergic airway inflammation via the rapid upregulation of MAPK phosphatase (MKP)‐1, which functions as a negative regulator of inflammation by deactivating p38 and JNK mitogen‐activated protein kinases (MAPKs). However, the role of endogenous Gln remains to be elucidated. Therefore, we investigated the mechanism by which endogenous Gln regulates MKP‐1 induction and allergic airway inflammation in an ovalbumin‐based murine asthma model. Methods We depleted endogenous Gln levels using L‐γ‐glutamyl‐p‐nitroanilide (GPNA), an inhibitor of the Gln transporter ASCT2 and glutamine synthetase small interfering siRNA. Lentivirus expressing MKP‐1 was injected to achieve overexpression of MKP‐1. Asthmatic phenotypes were assessed using our previously developed ovalbumin‐based murine model, which is suitable for examining sequential asthmatic events, including neutrophil infiltration. Gln levels were analyzed using a Gln assay kit. Results GPNA or glutamine synthetase siRNA successfully depleted endogenous Gln levels. Importantly, homeostatic MKP‐1 induction did not occur at all, which resulted in prolonged p38 MAPK and cytosolic phospholipase A2 (cPLA2) phosphorylation in Gln‐deficient mice. Gln deficiency augmented all examined asthmatic reactions, but it exhibited a strong bias toward increasing the neutrophil count, which was not observed in MKP‐1‐overexpressing lungs. This neutrophilia was inhibited by a cPLA2 inhibitor and a leukotriene B4 inhibitor but not by dexamethasone. Conclusion Gln deficiency leads to the impairment of MKP‐1 induction and activation of p38 MAPK and cPLA2, resulting in the augmentation of neutrophilic, more so than eosinophilic, airway inflammation.
Collapse
Affiliation(s)
- June‐Mo Kim
- Department of Immunology and Institute for Medical Science Jeonbuk National University Medical School Jeonju South Korea
| | - Yoo Na Im
- Department of Immunology and Institute for Medical Science Jeonbuk National University Medical School Jeonju South Korea
| | - Yun‐Jo Chung
- Center for University‐wide Research Facilities Jeonbuk National University Medical School Jeonju South Korea
| | - Jung‐ho Youm
- Department of Preventive Medicine Jeonbuk National University Medical School Jeonju South Korea
| | - Suhn Young Im
- Department of Biological Sciences College of Natural Sciences Chonnam National University Gwangju South Korea
| | - Myung Kwan Han
- Department of Microbiology and Institute for Medical Science Jeonbuk National University Medical School Jeonju South Korea
| | - Hern Ku Lee
- Department of Immunology and Institute for Medical Science Jeonbuk National University Medical School Jeonju South Korea
| |
Collapse
|
108
|
Differences and similarities in endothelial and angiogenic profiles of preeclampsia and COVID-19 in pregnancy. Am J Obstet Gynecol 2022; 227:277.e1-277.e16. [PMID: 35351411 PMCID: PMC8957356 DOI: 10.1016/j.ajog.2022.03.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022]
Abstract
Background COVID-19 presents a spectrum of signs and symptoms in pregnant women that might resemble preeclampsia. Differentiation between severe COVID-19 and preeclampsia is difficult in some cases. Objective To study biomarkers of endothelial damage, coagulation, innate immune response, and angiogenesis in preeclampsia and COVID-19 in pregnancy in addition to in vitro alterations in endothelial cells exposed to sera from pregnant women with preeclampsia and COVID-19. Study Design Plasma and sera samples were obtained from pregnant women with COVID-19 infection classified into mild (n=10) or severe (n=9) and from women with normotensive pregnancies as controls (n=10) and patients with preeclampsia (n=13). A panel of plasmatic biomarkers was assessed, including vascular cell adhesion molecule-1, soluble tumor necrosis factor-receptor I, heparan sulfate, von Willebrand factor antigen (activity and multimeric pattern), α2-antiplasmin, C5b9, neutrophil extracellular traps, placental growth factor, soluble fms-like tyrosine kinase-1, and angiopoietin 2. In addition, microvascular endothelial cells were exposed to patients’ sera, and changes in the cell expression of intercellular adhesion molecule 1 on cell membranes and von Willebrand factor release to the extracellular matrix were evaluated through immunofluorescence. Changes in inflammation cell signaling pathways were also assessed by of p38 mitogen-activated protein kinase phosphorylation. Statistical analysis included univariate and multivariate methods. Results Biomarker profiles of patients with mild COVID-19 were similar to those of controls. Both preeclampsia and severe COVID-19 showed significant alterations in most circulating biomarkers with distinctive profiles. Whereas severe COVID-19 exhibited higher concentrations of vascular cell adhesion molecule-1, soluble tumor necrosis factor-α receptor I, heparan sulfate, von Willebrand factor antigen, and neutrophil extracellular traps, with a significant reduction of placental growth factor compared with controls, preeclampsia presented a marked increase in vascular cell adhesion molecule-1 and soluble tumor necrosis factor-α receptor I (significantly increased compared with controls and patients with severe COVID-19), with a striking reduction in von Willebrand factor antigen, von Willebrand factor activity, and α2-antiplasmin. As expected, reduced placental growth factor, increased soluble fms-like tyrosine kinase-1 and angiopoietin 2, and a very high soluble fms-like tyrosine kinase-1 to placental growth factor ratio were also observed in preeclampsia. In addition, a significant increase in C5b9 and neutrophil extracellular traps was also detected in preeclampsia compared with controls. Principal component analysis demonstrated a clear separation between patients with preeclampsia and the other groups (first and second components explained 42.2% and 13.5% of the variance), mainly differentiated by variables related to von Willebrand factor, soluble tumor necrosis factor-receptor I, heparan sulfate, and soluble fms-like tyrosine kinase-1. Von Willebrand factor multimeric analysis revealed the absence of von Willebrand factor high-molecular-weight multimers in preeclampsia (similar profile to von Willebrand disease type 2A), whereas in healthy pregnancies and COVID-19 patients, von Willebrand factor multimeric pattern was normal. Sera from both preeclampsia and severe COVID-19 patients induced an overexpression of intercellular adhesion molecule 1 and von Willebrand factor in endothelial cells in culture compared with controls. However, the effect of preeclampsia was less pronounced than the that of severe COVID-19. Immunoblots of lysates from endothelial cells exposed to mild and severe COVID-19 and preeclampsia sera showed an increase in p38 mitogen-activated protein kinase phosphorylation. Patients with severe COVID-19 and preeclampsia were statistically different from controls, suggesting that both severe COVID-19 and preeclampsia sera can activate inflammatory signaling pathways. Conclusion Although similar in in vitro endothelial dysfunction, preeclampsia and severe COVID-19 exhibit distinctive profiles of circulating biomarkers related to endothelial damage, coagulopathy, and angiogenic imbalance that could aid in the differential diagnosis of these entities.
Collapse
|
109
|
Multi-omics evaluation of SARS-CoV-2 infected mouse lungs reveals dynamics of host responses. iScience 2022; 25:103967. [PMID: 35224468 PMCID: PMC8863311 DOI: 10.1016/j.isci.2022.103967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/04/2022] [Accepted: 02/17/2022] [Indexed: 12/27/2022] Open
Abstract
The outbreak of Coronavirus disease 2019 (COVID-19) throughout the world has caused millions of death, while the dynamics of host responses and the underlying regulation mechanisms during SARS-CoV-2 infection are not well depicted. Lung tissues from a mouse model sensitized to SARS-CoV-2 infection were serially collected at different time points for evaluation of transcriptome, proteome, and phosphoproteome. We showed the ebb and flow of several host responses in the lung across the viral infection. The signaling pathways and kinases regulating networks were alternated at different phases of infection. This multiplex evaluation also revealed that many kinases of the CDK and MAPK family were interactive and served as functional hubs in mediating the signal transduction during SARS-CoV-2 infection. Our study not only revealed the dynamics of lung pathophysiology and their underlying molecular mechanisms during SARS-CoV-2 infection, but also highlighted some molecules and signaling pathways that might guide future investigations on COVID-19 therapies. Multi-omics analysis profiles temporal host responses in SARS-CoV-2 infected lungs Signaling pathways and kinase regulating networks are dynamically altered The CDK and MAPK family are interactive and involved in regulating host responses
Collapse
|
110
|
Ye H, Kang L, Yan X, Li S, Huang Y, Mu R, Duan X, Chen L. MiR-103a-3p Promotes Zika Virus Replication by Targeting OTU Deubiquitinase 4 to Activate p38 Mitogen-Activated Protein Kinase Signaling Pathway. Front Microbiol 2022; 13:862580. [PMID: 35317262 PMCID: PMC8934420 DOI: 10.3389/fmicb.2022.862580] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/18/2022] [Indexed: 12/21/2022] Open
Abstract
Background MicroRNAs (miRNAs) play critical roles in regulating virus infection and replication. However, the mechanism by which miRNA regulates Zika virus (ZIKV) replication remains elusive. We aim to explore how the differentially expressed miR-103a-3p regulates ZIKV replication and to clarify the underlying molecular mechanism. Methods Small RNA sequencing (RNA-Seq) was performed to identify differentially expressed miRNAs in A549 cells with or without ZIKV infection and some of the dysregulated miRNAs were validated by quantitative real time PCR (qRT-PCR). The effect of miR-103a-3p on ZIKV replication was examined by transfecting miR-103a-3p mimic or negative control (NC) into A549 cells with or without p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 and expression levels of ZIKV NS5 mRNA and NS1 protein were detected by qRT-PCR and Western blot, respectively. The potential target genes for miR-103a-3p were predicted by four algorithms and further validated by mutation analysis through luciferase reporter assay. The predicated target gene OTU deubiquitinase (DUB) 4 (OTUD4) was over-expressed by plasmid transfection or silenced by siRNA transfection into cells prior to ZIKV infection. Activation status of p38 MAPK signaling pathway was revealed by looking at the phosphorylation levels of p38 (p-p38) and HSP27 (p-HSP27) by Western blot. Results Thirty-five differentially expressed miRNAs in ZIKV-infected A549 cells were identified by RNA-Seq analysis. Five upregulated and five downregulated miRNAs were further validated by qRT-PCR. One of the validated upregulated miRNAs, miR-103a-3p significantly stimulated ZIKV replication both at mRNA (NS5) and protein (NS1) levels. We found p38 MAPK signaling was activated following ZIKV infection, as demonstrated by the increased expression of the phosphorylation of p38 MAPK and HSP27. Blocking p38 MAPK signaling pathway using SB203580 inhibited ZIKV replication and attenuated the stimulating effect of miR-103a-3p on ZIKV replication. We further identified OTUD4 as a direct target gene of miR-103a-3p. MiR-103a-3p over-expression or OTUD4 silencing activated p38 MAPK signaling and enhanced ZIKV replication. In contrast, OTUD4 over-expression inhibited p38 MAPK activation and decreased ZIKV replication. In addition, OTUD4 over-expression attenuated the stimulating effect of miR-103a-3p on ZIKV replication and activation of p38 MAPK signaling. Conclusion Zika virus infection induced the expression of miR-103a-3p, which subsequently activated p38 MAPK signaling pathway by targeting OTUD4 to facilitate ZIKV replication.
Collapse
Affiliation(s)
- Haiyan Ye
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Lan Kang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Xipeng Yan
- The Joint Laboratory on Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
| | - Shilin Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Yike Huang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Rongrong Mu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Xiaoqiong Duan
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
- *Correspondence: Xiaoqiong Duan,
| | - Limin Chen
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
- The Joint Laboratory on Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
- Limin Chen,
| |
Collapse
|
111
|
Tang Y, Li X, Yuan Y, Zhang H, Zou Y, Xu Z, Xu Q, Song J, Deng C, Wang Q. Network pharmacology-based predictions of active components and pharmacological mechanisms of Artemisia annua L. for the treatment of the novel Corona virus disease 2019 (COVID-19). BMC Complement Med Ther 2022; 22:56. [PMID: 35241045 PMCID: PMC8893058 DOI: 10.1186/s12906-022-03523-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/28/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Novel Corona Virus Disease 2019 (COVID-19) is closely associated with cytokines storms. The Chinese medicinal herb Artemisia annua L. (A. annua) has been traditionally used to control many inflammatory diseases, such as malaria and rheumatoid arthritis. We performed network analysis and employed molecular docking and network analysis to elucidate active components or targets and the underlying mechanisms of A. annua for the treatment of COVID-19. METHODS Active components of A. annua were identified through the TCMSP database according to their oral bioavailability (OB) and drug-likeness (DL). Moreover, target genes associated with COVID-19 were mined from GeneCards, OMIM, and TTD. A compound-target (C-T) network was constructed to predict the relationship of active components with the targets. A Compound-disease-target (C-D-T) network has been built to reveal the direct therapeutic target for COVID-19. Molecular docking, molecular dynamics simulation studies (MD), and MM-GBSA binding free energy calculations were used to the closest molecules and targets between A. annua and COVID-19. RESULTS In our network, GO, and KEGG analysis indicated that A. annua acted in response to COVID-19 by regulating inflammatory response, proliferation, differentiation, and apoptosis. The molecular docking results manifested excellent results to verify the binding capacity between the hub components and hub targets in COVID-19. MD and MM-GBSA data showed quercetin to be the more effective candidate against the virus by target MAPK1, and kaempferol to be the other more effective candidate against the virus by target TP53. We identified A. annua's potentially active compounds and targets associated with them that act against COVID-19. CONCLUSIONS These findings suggest that A. annua may prevent and inhibit the inflammatory processes related to COVID-19.
Collapse
Affiliation(s)
- Yexiao Tang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaobo Li
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Sci-tech Industrial Park, Guangzhou University of Chinese Medicine, Guangzhou, 510445, China
| | - Yueming Yuan
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Sci-tech Industrial Park, Guangzhou University of Chinese Medicine, Guangzhou, 510445, China
| | - Hongying Zhang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Sci-tech Industrial Park, Guangzhou University of Chinese Medicine, Guangzhou, 510445, China
| | - Yuanyuan Zou
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhiyong Xu
- Sci-tech Industrial Park, Guangzhou University of Chinese Medicine, Guangzhou, 510445, China
| | - Qin Xu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jianping Song
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Changsheng Deng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qi Wang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Guangzhou Chest Hospital, Guangzhou, 510095, China.
| |
Collapse
|
112
|
Ahern DJ, Ai Z, Ainsworth M, Allan C, Allcock A, Angus B, Ansari MA, Arancibia-Cárcamo CV, Aschenbrenner D, Attar M, Baillie JK, Barnes E, Bashford-Rogers R, Bashyal A, Beer S, Berridge G, Beveridge A, Bibi S, Bicanic T, Blackwell L, Bowness P, Brent A, Brown A, Broxholme J, Buck D, Burnham KL, Byrne H, Camara S, Candido Ferreira I, Charles P, Chen W, Chen YL, Chong A, Clutterbuck EA, Coles M, Conlon CP, Cornall R, Cribbs AP, Curion F, Davenport EE, Davidson N, Davis S, Dendrou CA, Dequaire J, Dib L, Docker J, Dold C, Dong T, Downes D, Drakesmith H, Dunachie SJ, Duncan DA, Eijsbouts C, Esnouf R, Espinosa A, Etherington R, Fairfax B, Fairhead R, Fang H, Fassih S, Felle S, Fernandez Mendoza M, Ferreira R, Fischer R, Foord T, Forrow A, Frater J, Fries A, Gallardo Sanchez V, Garner LC, Geeves C, Georgiou D, Godfrey L, Golubchik T, Gomez Vazquez M, Green A, Harper H, Harrington HA, Heilig R, Hester S, Hill J, Hinds C, Hird C, Ho LP, Hoekzema R, Hollis B, Hughes J, Hutton P, Jackson-Wood MA, Jainarayanan A, James-Bott A, Jansen K, Jeffery K, Jones E, Jostins L, Kerr G, Kim D, Klenerman P, Knight JC, Kumar V, et alAhern DJ, Ai Z, Ainsworth M, Allan C, Allcock A, Angus B, Ansari MA, Arancibia-Cárcamo CV, Aschenbrenner D, Attar M, Baillie JK, Barnes E, Bashford-Rogers R, Bashyal A, Beer S, Berridge G, Beveridge A, Bibi S, Bicanic T, Blackwell L, Bowness P, Brent A, Brown A, Broxholme J, Buck D, Burnham KL, Byrne H, Camara S, Candido Ferreira I, Charles P, Chen W, Chen YL, Chong A, Clutterbuck EA, Coles M, Conlon CP, Cornall R, Cribbs AP, Curion F, Davenport EE, Davidson N, Davis S, Dendrou CA, Dequaire J, Dib L, Docker J, Dold C, Dong T, Downes D, Drakesmith H, Dunachie SJ, Duncan DA, Eijsbouts C, Esnouf R, Espinosa A, Etherington R, Fairfax B, Fairhead R, Fang H, Fassih S, Felle S, Fernandez Mendoza M, Ferreira R, Fischer R, Foord T, Forrow A, Frater J, Fries A, Gallardo Sanchez V, Garner LC, Geeves C, Georgiou D, Godfrey L, Golubchik T, Gomez Vazquez M, Green A, Harper H, Harrington HA, Heilig R, Hester S, Hill J, Hinds C, Hird C, Ho LP, Hoekzema R, Hollis B, Hughes J, Hutton P, Jackson-Wood MA, Jainarayanan A, James-Bott A, Jansen K, Jeffery K, Jones E, Jostins L, Kerr G, Kim D, Klenerman P, Knight JC, Kumar V, Kumar Sharma P, Kurupati P, Kwok A, Lee A, Linder A, Lockett T, Lonie L, Lopopolo M, Lukoseviciute M, Luo J, Marinou S, Marsden B, Martinez J, Matthews PC, Mazurczyk M, McGowan S, McKechnie S, Mead A, Mentzer AJ, Mi Y, Monaco C, Montadon R, Napolitani G, Nassiri I, Novak A, O'Brien DP, O'Connor D, O'Donnell D, Ogg G, Overend L, Park I, Pavord I, Peng Y, Penkava F, Pereira Pinho M, Perez E, Pollard AJ, Powrie F, Psaila B, Quan TP, Repapi E, Revale S, Silva-Reyes L, Richard JB, Rich-Griffin C, Ritter T, Rollier CS, Rowland M, Ruehle F, Salio M, Sansom SN, Sanches Peres R, Santos Delgado A, Sauka-Spengler T, Schwessinger R, Scozzafava G, Screaton G, Seigal A, Semple MG, Sergeant M, Simoglou Karali C, Sims D, Skelly D, Slawinski H, Sobrinodiaz A, Sousos N, Stafford L, Stockdale L, Strickland M, Sumray O, Sun B, Taylor C, Taylor S, Taylor A, Thongjuea S, Thraves H, Todd JA, Tomic A, Tong O, Trebes A, Trzupek D, Tucci FA, Turtle L, Udalova I, Uhlig H, van Grinsven E, Vendrell I, Verheul M, Voda A, Wang G, Wang L, Wang D, Watkinson P, Watson R, Weinberger M, Whalley J, Witty L, Wray K, Xue L, Yeung HY, Yin Z, Young RK, Youngs J, Zhang P, Zurke YX. A blood atlas of COVID-19 defines hallmarks of disease severity and specificity. Cell 2022; 185:916-938.e58. [PMID: 35216673 PMCID: PMC8776501 DOI: 10.1016/j.cell.2022.01.012] [Show More Authors] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/16/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023]
Abstract
Treatment of severe COVID-19 is currently limited by clinical heterogeneity and incomplete description of specific immune biomarkers. We present here a comprehensive multi-omic blood atlas for patients with varying COVID-19 severity in an integrated comparison with influenza and sepsis patients versus healthy volunteers. We identify immune signatures and correlates of host response. Hallmarks of disease severity involved cells, their inflammatory mediators and networks, including progenitor cells and specific myeloid and lymphocyte subsets, features of the immune repertoire, acute phase response, metabolism, and coagulation. Persisting immune activation involving AP-1/p38MAPK was a specific feature of COVID-19. The plasma proteome enabled sub-phenotyping into patient clusters, predictive of severity and outcome. Systems-based integrative analyses including tensor and matrix decomposition of all modalities revealed feature groupings linked with severity and specificity compared to influenza and sepsis. Our approach and blood atlas will support future drug development, clinical trial design, and personalized medicine approaches for COVID-19.
Collapse
|
113
|
Liang X, Chen Y, Fan Y. Bioinformatics approach to identify common gene signatures of patients with coronavirus 2019 and lung adenocarcinoma. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:22012-22030. [PMID: 34775559 PMCID: PMC8590527 DOI: 10.1007/s11356-021-17321-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/28/2021] [Indexed: 02/05/2023]
Abstract
Coronavirus disease 2019 (COVID-19) continues as a global pandemic. Patients with lung cancer infected with COVID-19 may develop severe disease or die. Treating such patients severely burdens overwhelmed healthcare systems. Here, we identified potential pathological mechanisms shared between patients with COVID-19 and lung adenocarcinoma (LUAD). Co-expressed, differentially expressed genes (DEGs) in patients with COVID-19 and LUAD were identified and used to construct a protein-protein interaction (PPI) network and to perform enrichment analysis. We used the NetworkAnalyst platform to establish a co-regulatory of the co-expressed DEGs, and we used Spearman's correlation to evaluate the significance of associations of hub genes with immune infiltration and immune checkpoints. Analysis of three datasets identified 112 shared DEGs, which were used to construct a protein-PPI network. Subsequent enrichment analysis revealed co-expressed genes related to biological process (BP), molecular function (MF), and cellular component (CC) as well as to pathways, specific organs, cells, and diseases. Ten co-expressed hub genes were employed to construct a gene-miRNA, transcription factor (TF)-gene, and TF-miRNA network. Hub genes were significantly associated with immune infiltration and immune checkpoints. Finally, methylation level of hub genes in LUAD was obtained via UALCAN database. The present multi-dimensional study reveals commonality in specific gene expression by patients with COVID-19 and LUAD. These findings provide insights into developing strategies for optimising the management and treatment of patients with LUAD with COVID-19.
Collapse
Affiliation(s)
- Xiao Liang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yali Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuchao Fan
- Department of Anesthesiology, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, No.55, Section 4, Renmin South Road, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
114
|
Nakou E, De Garate E, Liang K, Williams M, Pennell DJ, Bucciarelli-Ducci C. Imaging Findings of COVID-19-Related Cardiovascular Complications. Card Electrophysiol Clin 2022; 14:79-93. [PMID: 35221088 PMCID: PMC8556547 DOI: 10.1016/j.ccep.2021.10.008] [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] [Indexed: 12/15/2022]
Abstract
Other than respiratory disease, patients with coronavirus disease 2019 (COVID-19) commonly have cardiovascular manifestations, which are recognized as significant risk factors for increased mortality. COVID-19 patients may present with a wide spectrum of clinical presentations ranging from asymptomatic heart disease detected incidentally by cardiac investigations (troponin, BNP, and imaging) to cardiogenic shock and sudden cardiac death. In this broad clinical course, advanced imaging plays an important role in the diagnosis of different patterns of myocardial injury, risk stratification of COVID-19 patients, and in detecting potential cardiac side effects of the current treatments and vaccines against the severe acute respiratory syndrome.
Collapse
Affiliation(s)
- Eleni Nakou
- CMR Unit, Royal Brompton and Harefield Hospitals, Guys and St Thomas NHS Foundation Trust, Sydney Street, London, SW3 6NP, UK
| | - Estefania De Garate
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Trust and University of Bristol, Upper Maudlin St, Bristol, BS2 8HW, UK
| | - Kate Liang
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Trust and University of Bristol, Upper Maudlin St, Bristol, BS2 8HW, UK
| | - Matthew Williams
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Trust and University of Bristol, Upper Maudlin St, Bristol, BS2 8HW, UK
| | - Dudley J Pennell
- Royal Brompton and Harefield Hospitals, Guys and St Thomas NHS Trust and Imperial College London, London, SW3 6NP, UK
| | - Chiara Bucciarelli-Ducci
- Royal Brompton and Harefield Hospitals, Guys and St Thomas NHS Trust and King's College London, Sydney Street, London, SW3 6NP, UK.
| |
Collapse
|
115
|
The Promising Mechanisms of Low Molecular Weight Compounds of Panax Ginseng C.A. Meyer in Alleviating COVID-19: A Network Pharmacology Analysis. Processes (Basel) 2022. [DOI: 10.3390/pr10020333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Panax Ginseng C.A. Meyer (PGCAM) is a well-known phytomedicine, but most of its compounds, such as ginsenoside derivatives, have poor absorption and bioavailability profile due to high molecular weight (≥500 Daltons), which is the major hurdle for their clinical application. Hence, this research explored the efficiency of low molecular weight compounds (LMWCs) (<500 Daltons) screened from PGCAM and their anti-COVID-19 mechanisms through network pharmacology. Molecular compounds from PGCAM were identified using public databases and filtered out by the drug-likeness evaluation. Genes interacted with these filtered compounds, and COVID-19-related genes were extracted from public databases. In addition, overlapping genes between compounds and interactive genes were identified using the Venn diagram. In parallel, the networking between compounds and overlapping genes was analyzed by RStudio. The pathway enrichment analysis of overlapping genes was determined by STRING. Finally, the key bioactive compounds were documented through virtual screening. The bubble chart suggested that the mechanisms of PGCAM against COVID-19 were related to 28 signaling pathways. The key molecular anti-COVID-19 mechanisms might be the anti-inflammation, anti-permeability, and pro-apoptosis by inactivating the PI3K-Akt signaling pathway. The six key genes and the five compounds related to the PI3K-Akt signaling pathway were RELA-paeonol, NFKB1-frutinone A, IL6-nepetin, MCL1-ramalic acid, VEGFA-trifolirhizin, and IL2-trifolirhizin. The docking between these key genes and compounds demonstrated promising binding affinity with a good binding score. Overall, our proposed LMWCs from PGCAM provide a fundamental basis with noteworthy pharmacological evidence to support the therapeutic efficacy of PGCAM in relieving the main symptoms of COVID-19.
Collapse
|
116
|
Wan Afifudeen CL, Teh KY, Cha TS. Bioprospecting of microalgae metabolites against cytokine storm syndrome during COVID-19. Mol Biol Rep 2022; 49:1475-1490. [PMID: 34751914 PMCID: PMC8576090 DOI: 10.1007/s11033-021-06903-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 10/29/2021] [Indexed: 01/18/2023]
Abstract
In viral respiratory infections, disrupted pathophysiological outcomes have been attributed to hyper-activated and unresolved inflammation responses of the immune system. Integration between available drugs and natural therapeutics have reported benefits in relieving inflammation-related physiological outcomes and microalgae may be a feasible source from which to draw from against future coronavirus-infections. Microalgae represent a large and diverse source of chemically functional compounds such as carotenoids and lipids that possess various bioactivities, including anti-inflammatory properties. Therefore in this paper, some implicated pathways causing inflammation in viral respiratory infections are discussed and juxtaposed along with available research done on several microalgal metabolites. Additionally, the therapeutic properties of some known anti-inflammatory, antioxidant and immunomodulating compounds sourced from microalgae are reported for added clarity.
Collapse
Affiliation(s)
- Che Lah Wan Afifudeen
- Satreps-Cosmos Laboratory, Central Laboratory Complex, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu Malaysia
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu Malaysia
| | - Kit Yinn Teh
- Satreps-Cosmos Laboratory, Central Laboratory Complex, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu Malaysia
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu Malaysia
| | - Thye San Cha
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu Malaysia
- Satreps-Cosmos Laboratory, Central Laboratory Complex, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu Malaysia
| |
Collapse
|
117
|
Pu J, Chen D, Tian G, He J, Huang Z, Zheng P, Mao X, Yu J, Luo J, Luo Y, Yan H, Yu B. All-Trans Retinoic Acid Attenuates Transmissible Gastroenteritis Virus-Induced Inflammation in IPEC-J2 Cells via Suppressing the RLRs/NF-κB Signaling Pathway. Front Immunol 2022; 13:734171. [PMID: 35173714 PMCID: PMC8841732 DOI: 10.3389/fimmu.2022.734171] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/06/2022] [Indexed: 01/03/2023] Open
Abstract
Transmissible gastroenteritis virus (TGEV) infection can cause transmissible gastroenteritis (TGE), especially in suckling piglets, resulting in a significant economic loss for the global pig industry. The pathogenesis of TGEV infection is closely related to intestinal inflammation. All-trans retinoic acid (ATRA) has anti-inflammatory activity and immunomodulatory properties, but it is unclear whether ATRA can attenuate the inflammatory response induced by TGEV. This study aimed to investigate the protective effect of ATRA on TGEV-induced inflammatory injury in intestinal porcine epithelial cells (IPEC-J2) and to explore the underlying molecular mechanism. The results showed that TGEV infection triggered inflammatory response and damaged epithelial barrier integrity in IPEC-J2 cells. However, ATRA attenuated TGEV-induced inflammatory response by inhibiting the release of pro-inflammatory cytokines, including IL-1β, IL-6, IL-8 and TNF-α. ATRA also significantly reversed the reduction of ZO-1 and Occludin protein levels induced by TGEV infection and maintained epithelial barrier integrity. Moreover, ATRA treatment significantly prevented the upregulation of IкBα and NF-κB p65 phosphorylation levels and the nuclear translocation of NF-кB p65 induced by TGEV. On the other hand, treatment of TGEV-infected IPEC-J2 cells with the NF-κB inhibitors (BAY11-7082) significantly decreased the levels of inflammatory cytokines. Furthermore, ATRA treatment significantly downregulated the mRNA abundance and protein levels of TLR3, TLR7, RIG-I and MDA5, and downregulated their downstream signaling molecules TRIF, TRAF6 and MAVS mRNA expressions in TGEV-infected IPEC-J2 cells. However, the knockdown of RIG-I and MDA5 but not TLR3 and TLR7 significantly reduced the NF-κB p65 phosphorylation level and inflammatory cytokines levels in TGEV-infected IPEC-J2 cells. Our results indicated that ATRA attenuated TGEV-induced IPEC-J2 cells damage via suppressing inflammatory response, the mechanism of which is associated with the inhibition of TGEV-mediated activation of the RLRs/NF-κB signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education/Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
118
|
Liu R, Verma N, Henderson JA, Zhan S, Shen J. Profiling MAP kinase cysteines for targeted covalent inhibitor design. RSC Med Chem 2022; 13:54-63. [PMID: 35224496 PMCID: PMC8792824 DOI: 10.1039/d1md00277e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/28/2021] [Indexed: 07/20/2023] Open
Abstract
Mitogen-activated protein kinases (MAPK) are important therapeutic targets, and yet no inhibitors have advanced to the market. Here we applied the GPU-accelerated continuous constant pH molecular dynamics (CpHMD) to calculate the pK a's and profile the cysteine reactivities of all 14 MAPKs for assisting the targeted covalent inhibitor design. The simulations not only recapitulated but also rationalized the reactive cysteines in the front pocket of JNK1/2/3 and the extended front pocket of p38α. Interestingly, the DFG - 1 cysteine in the DFG-in conformation of ERK1/ERK2 was found somewhat reactive or unreactive; however, simulations of MKK7 showed that switching to the DFG-out conformation makes the DFG - 1 cysteine reactive, suggesting the advantage of type II covalent inhibitors. Additionally, the simulations prospectively predicted several druggable cysteine and lysine sites, including the αH head cysteine in JNK1/3 and DFG + 6 cysteine in JNK2, corroborating the chemical proteomic screening data. Given the low cost and the ability to offer physics-based rationales, we envision CpHMD simulations to complement the chemo-proteomic platform for systematic profiling cysteine reactivities for targeted covalent drug discovery.
Collapse
Affiliation(s)
- Ruibin Liu
- University of Maryland School of Pharmacy Baltimore MD USA
| | - Neha Verma
- University of Maryland School of Pharmacy Baltimore MD USA
| | | | - Shaoqi Zhan
- University of Maryland School of Pharmacy Baltimore MD USA
| | - Jana Shen
- University of Maryland School of Pharmacy Baltimore MD USA
| |
Collapse
|
119
|
Abstract
Abstract
Viruses completely rely on the energy and metabolic systems of host cells for life activities. Viral infections usually lead to cytopathic effects and host diseases. To date, there are still no specific clinical vaccines or drugs against most viral infections. Therefore, understanding the molecular and cellular mechanisms of viral infections is of great significance to prevent and treat viral diseases. A variety of viral infections are related to the p38 MAPK signalling pathway, and p38 is an important host factor in virus-infected cells. Here, we introduce the different signalling pathways of p38 activation and then summarise how different viruses induce p38 phosphorylation. Finally, we provide a general summary of the effect of p38 activation on virus replication. Our review provides integrated data on p38 activation and viral infections and describes the potential application of targeting p38 as an antiviral strategy.
Collapse
|
120
|
Castro P, Palomo M, Moreno-Castaño AB, Fernández S, Torramadé-Moix S, Pascual G, Martinez-Sanchez J, Richardson E, Téllez A, Nicolas JM, Carreras E, Richardson PG, Badimon JJ, Escolar G, Diaz-Ricart M. Is the Endothelium the Missing Link in the Pathophysiology and Treatment of COVID-19 Complications? Cardiovasc Drugs Ther 2022; 36:547-560. [PMID: 34097193 PMCID: PMC8181544 DOI: 10.1007/s10557-021-07207-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 02/08/2023]
Abstract
Patients with COVID-19 present a wide spectrum of disease severity, from asymptomatic cases in the majority to serious disease leading to critical care and even death. Clinically, four different scenarios occur within the typical disease timeline: first, an incubation and asymptomatic period; second, a stage with mild symptoms due mainly to the virus itself; third, in up to 20% of the patients, a stage with severe symptoms where a hyperinflammatory response with a cytokine storm driven by host immunity induces acute respiratory distress syndrome; and finally, a post-acute sequelae (PASC) phase, which present symptoms that can range from mild or annoying to actually quite incapacitating. Although the most common manifestation is acute respiratory failure of the lungs, other organs are also frequently involved. The clinical manifestations of the COVID-19 infection support a key role for endothelial dysfunction in the pathobiology of this condition. The virus enters into the organism via its interaction with angiotensin-converting enzyme 2-receptor that is present prominently in the alveoli, but also in endothelial cells, which can be directly infected by the virus. Cytokine release syndrome can also drive endothelial damage independently. Consequently, a distinctive feature of SARS-CoV-2 infection is vascular harm, with severe endothelial injury, widespread thrombosis, microangiopathy, and neo-angiogenesis in response to endothelial damage. Therefore, endothelial dysfunction seems to be the pathophysiological substrate for severe COVID-19 complications. Biomarkers of endothelial injury could constitute strong indicators of disease progression and severity. In addition, the endothelium could represent a very attractive target to both prevent and treat these complications. To establish an adequate therapy, the underlying pathophysiology and corresponding clinical stage should be clearly identified. In this review, the clinical features of COVID-19, the central role of the endothelium in COVID-19 and in other pathologies, and the potential of specific therapies aimed at protecting the endothelium in COVID-19 patients are addressed.
Collapse
Affiliation(s)
- Pedro Castro
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Marta Palomo
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Ana Belen Moreno-Castaño
- Barcelona Endothelium Team, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | - Sara Fernández
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Sergi Torramadé-Moix
- IDIBAPS, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | | | - Julia Martinez-Sanchez
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Edward Richardson
- Frank H. Netter M.D. School of Medicine At, Quinnipiac University, North Haven, CT, USA
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Adrián Téllez
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Josep M Nicolas
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Enric Carreras
- Josep Carreras Leukaemia Research Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Paul G Richardson
- Jerome Lipper Multiple Myeloma Center, Division of Hematologic Malignancy, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Juan José Badimon
- Cardiology Department, Cardiovascular Institute, Mount Sinai Hospital, New York, NY, USA
- AtheroThrombosis Research Unit, Cardiovascular Institute, Icahn School of Medicine At Mount Sinai, New York, NY, USA
| | - Gines Escolar
- School of Medicine, University of Barcelona, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain
| | - Maribel Diaz-Ricart
- School of Medicine, University of Barcelona, Barcelona, Spain.
- IDIBAPS, Barcelona, Spain.
- Barcelona Endothelium Team, Barcelona, Spain.
- Hematopathology, Pathology Department, CDB, Hospital Clinic, Villarroel 170, 08036, Barcelona, Spain.
| |
Collapse
|
121
|
Liu Y, Kuang D, Li D, Yang J, Yan J, Xia Y, Zhang F, Cao H. Roles of the gut microbiota in severe SARS-CoV-2 infection. Cytokine Growth Factor Rev 2022; 63:98-107. [PMID: 35131164 PMCID: PMC8802135 DOI: 10.1016/j.cytogfr.2022.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/11/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread worldwide. The pathophysiological mechanisms linking gut dysbiosis and severe SARS-CoV-2 infection are poorly understood, although gut microbiota disorders are related to severe SARS-CoV-2 infections. The roles of the gut microbiota in severe SARS-CoV-2 infection were compared with those in respiratory viral infection, which is an easily understood and enlightening analogy. Secondary bacterial infections caused by immune disorders and antibiotic abuse can lead to dysregulation of the gut microbiota in patients with respiratory viral infections. The gut microbiota can influence the progression of respiratory viral infections through metabolites and the immune response, which is known as the gut–lung axis. Angiotensin-converting enzyme 2 is expressed in both the lungs and the small intestine, which may be a bridge between the lung and the gut. Similarly, SARS-CoV-2 infection has been shown to disturb the gut microbiota, which may be the cause of cytokine storms. Bacteria in the gut, lung, and other tissues and respiratory viruses can be considered microecosystems and may exert overall effects on the host. By referencing respiratory viral infections, this review focused on the mechanisms involved in the interaction between SARS-CoV-2 infections and the gut microbiota and provides new strategies for the treatment or prevention of severe SARS-CoV-2 infections by improving gut microbial homeostasis.
Collapse
|
122
|
Farahani M, Niknam Z, Mohammadi Amirabad L, Amiri-Dashatan N, Koushki M, Nemati M, Danesh Pouya F, Rezaei-Tavirani M, Rasmi Y, Tayebi L. Molecular pathways involved in COVID-19 and potential pathway-based therapeutic targets. Biomed Pharmacother 2022; 145:112420. [PMID: 34801852 PMCID: PMC8585639 DOI: 10.1016/j.biopha.2021.112420] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 01/08/2023] Open
Abstract
Deciphering the molecular downstream consequences of severe acute respiratory syndrome coronavirus (SARS-CoV)- 2 infection is important for a greater understanding of the disease and treatment planning. Furthermore, greater understanding of the underlying mechanisms of diagnostic and therapeutic strategies can help in the development of vaccines and drugs against COVID-19. At present, the molecular mechanisms of SARS-CoV-2 in the host cells are not sufficiently comprehended. Some of the mechanisms are proposed considering the existing similarities between SARS-CoV-2 and the other members of the β-CoVs, and others are explained based on studies advanced in the structure and function of SARS-CoV-2. In this review, we endeavored to map the possible mechanisms of the host response following SARS-CoV-2 infection and surveyed current research conducted by in vitro, in vivo and human observations, as well as existing suggestions. We addressed the specific signaling events that can cause cytokine storm and demonstrated three forms of cell death signaling following virus infection, including apoptosis, pyroptosis, and necroptosis. Given the elicited signaling pathways, we introduced possible pathway-based therapeutic targets; ADAM17 was especially highlighted as one of the most important elements of several signaling pathways involved in the immunopathogenesis of COVID-19. We also provided the possible drug candidates against these targets. Moreover, the cytokine-cytokine receptor interaction pathway was found as one of the important cross-talk pathways through a pathway-pathway interaction analysis for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Masoumeh Farahani
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Niknam
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nasrin Amiri-Dashatan
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehdi Koushki
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Nemati
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Fahima Danesh Pouya
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran; Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| |
Collapse
|
123
|
Alves HR, Lomba GSB, Gonçalves-de-Albuquerque CF, Burth P. Irisin, Exercise, and COVID-19. Front Endocrinol (Lausanne) 2022; 13:879066. [PMID: 35784579 PMCID: PMC9248970 DOI: 10.3389/fendo.2022.879066] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Muscle and adipose tissue produce irisin during exercise. Irisin is thermogenic adipomyokine, improves glucose and lipid metabolism, and ameliorates the effects of obesity-driven inflammation, metabolic syndrome, and diabetes. In addition, exercise-induced irisin activates anti-inflammatory pathways and may play an essential role in improving the outcomes of inflammatory conditions, such as coronavirus disease (COVID-19). COVID-19 infection can activate different intracellular receptors and modulate various pathways during the course of the disease. The cytokine release storm (CRS) produced is significant because it promotes the context for systemic inflammation, which increases the risk of mortality in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV2). In addition, viral infection and the resulting organ damage may stimulate the mitogen-activated protein kinase(MAPK) and toll-like receptor 4 (TLR4)/toll interleukin receptor (TIR)-domain-containing adaptor (MyD88) pathways while negatively modulating the AMP-activated protein kinase (AMPK) pathway, leading to increased inflammatory cytokine production. Exercise-induced irisin may counteract this inflammatory modulation by decreasing cytokine production. Consequently, increased irisin levels, as found in healthy patients, may favor a better prognosis in patients with SARS-CoV2. This review aims to explore the molecular mechanisms underlying the anti-inflammatory properties of irisin in mitigating CRS and preventing severe outcomes due to infection with SARS-CoV2.
Collapse
Affiliation(s)
- Hugo Rodrigues Alves
- Department of Cell and Molecular Biology, Fluminense Federal University, Niterói, Brazil
| | | | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate Program in Biotechnology, Fluminense Federal University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Burth, ; Cassiano Felippe Gonçalves-de-Albuquerque,
| | - Patricia Burth
- Department of Cell and Molecular Biology, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program in Biotechnology, Fluminense Federal University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Burth, ; Cassiano Felippe Gonçalves-de-Albuquerque,
| |
Collapse
|
124
|
Bi Z, Hong W, Que H, He C, Ren W, Yang J, Lu T, Chen L, Lu S, Peng X, Wei X. Inactivated SARS-CoV-2 induces acute respiratory distress syndrome in human ACE2-transgenic mice. Signal Transduct Target Ther 2021; 6:439. [PMID: 34952899 PMCID: PMC8705082 DOI: 10.1038/s41392-021-00851-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 02/07/2023] Open
Abstract
The development of animal models for COVID-19 is essential for basic research and drug/vaccine screening. Previously reported COVID-19 animal models need to be established under a high biosafety level condition for the utilization of live SARS-CoV-2, which greatly limits its application in routine research. Here, we generate a mouse model of COVID-19 under a general laboratory condition that captures multiple characteristics of SARS-CoV-2-induced acute respiratory distress syndrome (ARDS) observed in humans. Briefly, human ACE2-transgenic (hACE2) mice were intratracheally instilled with the formaldehyde-inactivated SARS-CoV-2, resulting in a rapid weight loss and detrimental changes in lung structure and function. The pulmonary pathologic changes were characterized by diffuse alveolar damage with pulmonary consolidation, hemorrhage, necrotic debris, and hyaline membrane formation. The production of fatal cytokines (IL-1β, TNF-α, and IL-6) and the infiltration of activated neutrophils, inflammatory monocyte-macrophages, and T cells in the lung were also determined, suggesting the activation of an adaptive immune response. Therapeutic strategies, such as dexamethasone or passive antibody therapy, could effectively ameliorate the disease progression in this model. Therefore, the established mouse model for SARS-CoV-2-induced ARDS in the current study may provide a robust tool for researchers in the standard open laboratory to investigate the pathological mechanisms or develop new therapeutic strategies for COVID-19 and ARDS.
Collapse
Affiliation(s)
- Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Wenyan Ren
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Tianqi Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Shuaiyao Lu
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China.
| | - Xiaozhong Peng
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China.
| |
Collapse
|
125
|
Lin C, Tsai FJ, Hsu YM, Ho TJ, Wang GK, Chiu YJ, Ha HA, Yang JS. Study of Baicalin toward COVID-19 Treatment: In silico Target Analysis and in vitro Inhibitory Effects on SARS-CoV-2 Proteases. Biomed Hub 2021; 6:122-137. [PMID: 34934765 PMCID: PMC8647113 DOI: 10.1159/000519564] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/02/2021] [Indexed: 01/18/2023] Open
Abstract
Negative impacts of COVID-19 on human health and economic and social activities urge scientists to develop effective treatments. Baicalin is a natural flavonoid, extracted from a traditional medicinal plant, previously reported with anti-inflammatory activity. In this study, we used pharmacophore fitting and molecular docking to screen and determine docking patterns and the binding affinity of baicalin on 3 major targets of SARS-CoV-2 (3-chymotrypsin-like cysteine protease [3CLpro], papain-like protease [PLpro], and RNA-dependent RNA polymerase). The obtained data revealed that baicalin has high pharmacophore fitting on 3CLpro and predicted good binding affinity on PLpro. Moreover, using the enzymatic assay, we examined the inhibitory effect of baicalin in vitro on the screened enzymes. Baicalin also exhibits inhibitory effect on these proteases in vitro. Additionally, we performed pharmacophore-based screening of baicalin on human targets and conducted pathway analysis to explore the potential cytoprotective effects of baicalin in the host cell that may be beneficial for COVID-19 treatment. The result suggested that baicalin has multiple targets in human cell that may induce multiple pharmacological effects. The result of pathway analysis implied that these targets may be associated with baicalin-induced bioactivities that are involved with signals of pro-inflammation factors, such as cytokine and chemokine. Taken together with supportive data from the literature, the bioactivities of bailalin may be beneficial for COVID-19 treatment by reducing cytokine-induced acute inflammation. In conclusion, baicalin is potentially a good candidate for developing new therapeutic to treat COVID-19.
Collapse
Affiliation(s)
- Chingju Lin
- Department of Physiology, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Medical Research, Human Genetics Center, China Medical University Hospital 404, Taichung, Taiwan.,Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Guo-Kai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, China
| | - Yu-Jen Chiu
- Division of Reconstructive and Plastic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, National Yang Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hai-Anh Ha
- School of Pharmacy, China Medical University, Taichung, Taiwan.,Faculty of Pharmacy, Duy Tan University, Da Nang, Vietnam
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
126
|
Identification of a dual acting SARS-CoV-2 proteases inhibitor through in silico design and step-by-step biological characterization. Eur J Med Chem 2021; 226:113863. [PMID: 34571172 PMCID: PMC8457654 DOI: 10.1016/j.ejmech.2021.113863] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/12/2022]
Abstract
COVID-19 pandemic, starting from the latest 2019, and caused by SARS-CoV-2 pathogen, led to the hardest health-socio-economic disaster in the last century. Despite the tremendous scientific efforts, mainly focused on the development of vaccines, identification of potent and efficient anti-SARS-CoV-2 therapeutics still represents an unmet need. Remdesivir, an anti-Ebola drug selected from a repurposing campaign, is the only drug approved, so far, for the treatment of the infection. Nevertheless, WHO in later 2020 has recommended against its use in COVID-19. In the present paper, we describe a step-by-step in silico design of a small library of compounds as main protease (Mpro) inhibitors. All the molecules were screened by an enzymatic assay on Mpro and, then, cellular activity was evaluated using Vero cells viral infection model. The cellular screening disclosed compounds 29 and 34 as in-vitro SARS-CoV-2 replication inhibitors at non-toxic concentrations (0.32 < EC50 < 5.98 μM). To rationalize these results, additional in-vitro assays were performed, focusing on papain like protease (PLpro) and spike protein (SP) as potential targets for the synthesized molecules. This pharmacological workflow allowed the identification of compound 29, as a dual acting SARS-CoV-2 proteases inhibitor featuring micromolar inhibitory potency versus Mpro (IC50 = 1.72 μM) and submicromolar potency versus PLpro (IC50 = 0.67 μM), and of compound 34 as a selective SP inhibitor (IC50 = 3.26 μM).
Collapse
|
127
|
Su C, Rousseau S, Emad A. Identification of transcriptional regulatory network associated with response of host epithelial cells to SARS-CoV-2. Sci Rep 2021; 11:23928. [PMID: 34907210 PMCID: PMC8671548 DOI: 10.1038/s41598-021-03309-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Identification of transcriptional regulatory mechanisms and signaling networks involved in the response of host cells to infection by SARS-CoV-2 is a powerful approach that provides a systems biology view of gene expression programs involved in COVID-19 and may enable the identification of novel therapeutic targets and strategies to mitigate the impact of this disease. In this study, our goal was to identify a transcriptional regulatory network that is associated with gene expression changes between samples infected by SARS-CoV-2 and those that are infected by other respiratory viruses to narrow the results on those enriched or specific to SARS-CoV-2. We combined a series of recently developed computational tools to identify transcriptional regulatory mechanisms involved in the response of epithelial cells to infection by SARS-CoV-2, and particularly regulatory mechanisms that are specific to this virus when compared to other viruses. In addition, using network-guided analyses, we identified kinases associated with this network. The results identified pathways associated with regulation of inflammation (MAPK14) and immunity (BTK, MBX) that may contribute to exacerbate organ damage linked with complications of COVID-19. The regulatory network identified herein reflects a combination of known hits and novel candidate pathways supporting the novel computational pipeline presented herein to quickly narrow down promising avenues of investigation when facing an emerging and novel disease such as COVID-19.
Collapse
Affiliation(s)
- Chen Su
- Department of Electrical and Computer Engineering, McGill University, 755, McConnell Engineering Building, 3480 University Street, Montreal, QC, H3A 0E9, Canada
| | - Simon Rousseau
- The Meakins-Christie Laboratories at the Research Institute of McGill University Heath Centre (RI-MUHC), McGill University, E M3.2244, 1001 Décarie, Montreal, QC, H4A 3J1, Canada.
- Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, 755, McConnell Engineering Building, 3480 University Street, Montreal, QC, H3A 0E9, Canada.
- The Meakins-Christie Laboratories at the Research Institute of McGill University Heath Centre (RI-MUHC), McGill University, E M3.2244, 1001 Décarie, Montreal, QC, H4A 3J1, Canada.
- Mila, Quebec AI Institute, Montreal, QC, Canada.
| |
Collapse
|
128
|
Comparisons of the immunological landscape of COVID-19 patients based on sex and disease severity by multi-omics analysis. Chem Biol Interact 2021; 352:109777. [PMID: 34896122 PMCID: PMC8654707 DOI: 10.1016/j.cbi.2021.109777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/28/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022]
Abstract
Objective To determine the differences in the immune response against SARS-CoV-2 infection of patients based on sex and disease severity. Methods We used an analytical framework of 382 transcriptional modules and multi-omics analyses to discriminate COVID-19 patients based on sex and disease severity. Results Male and female patients overexpressed modules related to the innate immune response. The expression of modules related to the adaptive immune response showed lower enrichment levels in males than females. Inflammation modules showed ascending overexpression in male and female patients, while a higher level was observed in severe female patients. Moderate female patients demonstrated significant overexpression to interferon, cytolytic lymphocyte, T & B cells, and erythrocytes modules. Moderate female patients showed a higher adaptive immune response than males matched group. Pathways involved in metabolism dysregulation and Hippo signaling were upregulated in females than in male patients. Females and moderate cases showed higher levels of metabolic dysregulation. Conclusions The immune landscape in COVID-19 patients was noticeably different between the sexes, and these differences may highlight disease vulnerability in males. This study suggested that certain treatments that increase or decrease the immune responses to SARS-CoV-2 might be necessary for male and female patients at certain disease stages.
Collapse
|
129
|
Abstract
COVID-19 has become a major pandemic in recent times. The exact pathophysiology and understanding of cytokine storm and immunomodulation are evolving. Various cytokines have been implicated in the pathophysiology of COVID-19. Immunosuppressant immunomodulators like steroids, canakinumab, anakinra, tocilizumab, sarilumab, baricitinib, ruxolitinib, bevacizumab, and itolizumab have been tried. Immunostimulant immunomodulators like interferons (IFNs) and Mycobacterium w (Mw) have also been repurposed. Considering the role of multiple cytokines implicated in COVID-19, molecules working on the majority of the targets, may hold a promising future prospect
Collapse
Affiliation(s)
- Pradeep Rangappa
- Department of Critical Care Medicine, Columbia Asia, Bengaluru, Karnataka, India
| |
Collapse
|
130
|
Herold R, Scholtysik R, Moroniak S, Weiss C, Ishikawa H, Schroten H, Schwerk C. Capsule-dependent impact of MAPK signalling on host cell invasion and immune response during infection of the choroid plexus epithelium by Neisseria meningitidis. Fluids Barriers CNS 2021; 18:53. [PMID: 34863201 PMCID: PMC8643193 DOI: 10.1186/s12987-021-00288-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/16/2021] [Indexed: 01/15/2023] Open
Abstract
Background The Gram-negative bacterium Neisseria meningitidis (Nm) can cause meningitis in humans, but the host signalling pathways manipulated by Nm during central nervous system (CNS) entry are not completely understood. Methods We investigate the role of the mitogen-activated protein kinases (MAPK) Erk1/2 and p38 in an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB) based on human epithelial choroid plexus (CP) papilloma (HIBCPP) cells during infection with Nm serogroup B (NmB) and serogroup C (NmC) strains. A transcriptome analysis of HIBCPP cells following infection with Nm by massive analysis of cDNA ends (MACE) was done to further characterize the cellular response to infection of the barrier. Results Interestingly, whereas NmB and NmC wild type strains required active Erk1/2 and p38 pathways for infection, invasion by capsule-deficient mutants was independent of Erk1/2 and, in case of the NmB strain, of p38 activity. The transcriptome analysis of HIBCPP cells following infection with Nm demonstrated specific regulation of genes involved in the immune response dependent on Erk1/2 signalling. Gene ontology (GO) analysis confirmed loss of MAPK signalling after Erk1/2 inhibition and revealed an additional reduction of cellular responses including NFκB and JAK-STAT signalling. Interestingly, GO terms related to TNF signalling and production of IL6 were lost specifically following Erk1/2 inhibition during infection with wild type Nm, which correlated with the reduced infection rates by the wild type in absence of Erk1/2 signalling. Conclusion Our data point towards a role of MAPK signalling during infection of the CP epithelium by Nm, which is strongly influenced by capsule expression, and affects infection rates as well as the host cell response. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-021-00288-7.
Collapse
Affiliation(s)
- Rosanna Herold
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - René Scholtysik
- Genomics & Transcriptomics Facility, Institute of Cell Biology, University Hospital Essen, Virchowstraße 173, 45122, Essen, Germany
| | - Selina Moroniak
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, 1-1-1Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| |
Collapse
|
131
|
Single-Cell and Bulk RNASeq Profiling of COVID-19 Patients Reveal Immune and Inflammatory Mechanisms of Infection-Induced Organ Damage. Viruses 2021; 13:v13122418. [PMID: 34960687 PMCID: PMC8706409 DOI: 10.3390/v13122418] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023] Open
Abstract
The SARS-CoV-2 virus’s ability to induce hypercytokinemia and cause multiple organ failure makes it imperative to find effective treatments. To understand the mechanism of viral infection and its effects on organ tissues, we analyzed multiple single-cell and bulk RNAseq data from COVID-19 patients’ organ samples. Various levels of severity of infection were accounted for, with comparative analyses between mild, moderate, and severely infected patients. Our analysis uncovered an upregulation of the innate immune response via several inflammatory genes, IL-2, IL-6, IL-8, IL-17A, and NF-κB. Consequently, we found that the upregulation of these downstream effects can lead to organ injury. The downregulated pathways such as eukaryotic initiation factor 2 (eIF2) and eIF4-mediated host translation, were found to lead to an increased viral translation. We also found that the loss of inhibitory peptides can suppress an overactive innate immune response via NF-κB and interleukin-mediated pathways. Investigation of viral-host protein mapping showed that the interaction of viral proteins with host proteins correlated with the down- and upregulation of host pathways such as decreased eIF2-mediated host translation and increased hypertrophy and fibrosis. Inflammation was increased via the stimulation of pro-inflammatory cytokines and suppression of host translation pathways that led to reduced inflammatory inhibitors. Cardiac hypertrophy and organ fibrosis were the results of increased inflammation in organs of severe and critical patients. Finally, we identified potential therapeutic targets for the treatment of COVID-19 and its deleterious effects on organs. Further experimental investigation would conclusively determine the effects of COVID-19 infection on organs other than the lungs and the effectiveness of the proposed therapeutic targets.
Collapse
|
132
|
Afroz S, Fairuz S, Joty JA, Uddin MN, Rahman MA. Virtual screening of functional foods and dissecting their roles in modulating gene functions to support post COVID-19 complications. J Food Biochem 2021; 45:e13961. [PMID: 34676581 PMCID: PMC8646449 DOI: 10.1111/jfbc.13961] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 12/19/2022]
Abstract
COVID-19 has become the focal point since 2019 after the outbreak of coronavirus disease. Many drugs are being tested and used to treat coronavirus infections; different kinds of vaccines are also introduced as preventive measure. Alternative therapeutics are as well incorporated into the health guidelines of some countries. This research aimed to look into the underlying mechanisms of functional foods and how they may improve the long-term post COVID-19 cardiovascular, diabetic, and respiratory complications through their bioactive compounds. The potentiality of nine functional foods for post COVID-19 complications was investigated through computational approaches. A total of 266 bioactive compounds of these foods were searched via extensive literature reviewing. Three highly associated targets namely troponin I interacting kinase (TNNI3K), dipeptidyl peptidase 4 (DPP-4), and transforming growth factor beta 1 (TGF-β1) were selected for cardiovascular, diabetes, and respiratory disorders, respectively, after COVID-19 infections. Best docked compounds were further analyzed by network pharmacological tools to explore their interactions with complication-related genes (MAPK1 and HSP90AA1 for cardiovascular, PPARG and TNF-alpha for diabetes, and AKT-1 for respiratory disorders). Seventy-one suggested compounds out of one-hundred and thirty-nine (139) docked compounds in network pharmacology recommended 169 Gene Ontology (GO) items and 99 Kyoto Encyclopedia of Genes and Genomes signaling pathways preferably AKT signaling pathway, MAPK signaling pathway, ACE2 receptor signaling pathway, insulin signaling pathway, and PPAR signaling pathway. Among the chosen functional foods, black cumin, fenugreek, garlic, ginger, turmeric, bitter melon, and Indian pennywort were found to modulate the actions. Results demonstrate that aforesaid functional foods have attenuating roles to manage post COVID-19 complications. PRACTICAL APPLICATIONS: Functional foods have been approaching a greater interest due to their medicinal uses other than gastronomic pleasure. Nine functional food resources have been used in this research for their traditional and ethnopharmacological uses, but their directive-role in modulating the genes involved in the management of post COVID-19 complications is inadequately studied and reported. Therefore, the foods types used in this research may be prioritized to be used as functional foods for ameliorating the major post COVID-19 complications through appropriate science.
Collapse
Affiliation(s)
- Sharmin Afroz
- Department of Theoretical and Computational ChemistryUniversity of DhakaDhakaBangladesh
| | | | - Jahanara Alam Joty
- Department of Biochemistry and BiotechnologyUniversity of Science and TechnologyChittagongBangladesh
| | - Md. Nazim Uddin
- Institute of Food Science and TechnologyBangladesh Council of Scientific and Industrial ResearchDhakaBangladesh
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| |
Collapse
|
133
|
Banerjee S, Yadav S, Banerjee S, Fakayode SO, Parvathareddy J, Reichard W, Surendranathan S, Mahmud F, Whatcott R, Thammathong J, Meibohm B, Miller DD, Jonsson CB, Dubey KD. Drug Repurposing to Identify Nilotinib as a Potential SARS-CoV-2 Main Protease Inhibitor: Insights from a Computational and In Vitro Study. J Chem Inf Model 2021; 61:5469-5483. [PMID: 34666487 PMCID: PMC8547516 DOI: 10.1021/acs.jcim.1c00524] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Indexed: 12/11/2022]
Abstract
COVID-19, an acute viral pneumonia, has emerged as a devastating pandemic. Drug repurposing allows researchers to find different indications of FDA-approved or investigational drugs. In this current study, a sequence of pharmacophore and molecular modeling-based screening against COVID-19 Mpro (PDB: 6LU7) suggested a subset of drugs, from the Drug Bank database, which may have antiviral activity. A total of 44 out of 8823 of the most promising virtual hits from the Drug Bank were subjected to molecular dynamics simulation experiments to explore the strength of their interactions with the SARS-CoV-2 Mpro active site. MD findings point toward three drugs (DB04020, DB12411, and DB11779) with very low relative free energies for SARS-CoV-2 Mpro with interactions at His41 and Met49. MD simulations identified an additional interaction with Glu166, which enhanced the binding affinity significantly. Therefore, Glu166 could be an interesting target for structure-based drug design. Quantitative structural-activity relationship analysis was performed on the 44 most promising hits from molecular docking-based virtual screening. Partial least square regression accurately predicted the values of independent drug candidates' binding energy with impressively high accuracy. Finally, the EC50 and CC50 of 10 drug candidates were measured against SARS-CoV-2 in cell culture. Nilotinib and bemcentinib had EC50 values of 2.6 and 1.1 μM, respectively. In summary, the results of our computer-aided drug design provide a roadmap for rational drug design of Mpro inhibitors and the discovery of certified medications as COVID-19 antiviral therapeutics.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Physical Sciences,
University of Arkansas Fort Smith, 5210 Grand Avenue, Fort
Smith, Arkansas 72904, United States
| | - Shalini Yadav
- Department of Chemistry, Shiv Nadar
University, Gautam Buddha Nagar, Uttar Pradesh 201314,
India
| | - Sourav Banerjee
- Department of Chemistry, School of Basic and Applied
Sciences, Adamas University, Kolkata 700126,
India
| | - Sayo O. Fakayode
- Department of Physical Sciences,
University of Arkansas Fort Smith, 5210 Grand Avenue, Fort
Smith, Arkansas 72904, United States
| | - Jyothi Parvathareddy
- Regional Biocontainment Laboratory,
University of Tennessee Health Science Center, 901 Monroe
Avenue, Memphis, Tennessee 38163, United States
| | - Walter Reichard
- Department of Microbiology, Immunology, and
Biochemistry, University of Tennessee Health Science Center,
Memphis, Tennessee 38163, United States
| | - Surekha Surendranathan
- Regional Biocontainment Laboratory,
University of Tennessee Health Science Center, 901 Monroe
Avenue, Memphis, Tennessee 38163, United States
| | - Foyez Mahmud
- Department of Bioengineering, Rice
University, 6100 Main Street, Houston, Texas 77005, United
States
| | - Ryan Whatcott
- Department of Physical Sciences,
University of Arkansas Fort Smith, 5210 Grand Avenue, Fort
Smith, Arkansas 72904, United States
| | - Joshua Thammathong
- Department of Physical Sciences,
University of Arkansas Fort Smith, 5210 Grand Avenue, Fort
Smith, Arkansas 72904, United States
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of
Pharmacy, University of Tennessee Health Science Center,
Memphis, Tennessee 38163, United States
| | - Duane D. Miller
- Department of Pharmaceutical Sciences, College of
Pharmacy, University of Tennessee Health Science Center,
Memphis, Tennessee 38163, United States
| | - Colleen B. Jonsson
- Regional Biocontainment Laboratory,
University of Tennessee Health Science Center, 901 Monroe
Avenue, Memphis, Tennessee 38163, United States
- Department of Microbiology, Immunology, and
Biochemistry, University of Tennessee Health Science Center,
Memphis, Tennessee 38163, United States
- Department of Pharmaceutical Sciences, College of
Pharmacy, University of Tennessee Health Science Center,
Memphis, Tennessee 38163, United States
| | - Kshatresh Dutta Dubey
- Department of Chemistry, Shiv Nadar
University, Gautam Buddha Nagar, Uttar Pradesh 201314,
India
| |
Collapse
|
134
|
Jia L, Cui W, Chen J, Yang J, Xue X, Cai J, Zhao W, Gao W. Erythropoietin alleviates acute lung injury induced by ischemia-reperfusion through blocking p38 MAPK signaling. Hum Exp Toxicol 2021; 40:S593-S602. [PMID: 34784828 DOI: 10.1177/09603271211043480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Erythropoietin (EPO) has antiapoptotic, antioxidative, and anti-inflammatory effects on ischemia tissues and protects against acute lung injury (ALI) induced by ischemia-reperfusion (I/R). p38 mitogen-activated protein kinases (p38 MAPK) signaling is involved in the processes of I/R-induced ALI. However, the interaction of EPO with p38 MAPK signaling in I/R-induced ALI has not been reported. To explore this issue, we constructed an I/R-induced ALI model in vivo and in vitro using Sprague Dawley rats and BEAS-2B cells. Some I/R rats and hypoxia-reoxygenation (H/R)-induced cells were treated with EPO, and the others were used as control groups. The injuries of lung tissues and cells were respectively assessed by inflammatory cytokine, morphologic changes, cell viability, apoptosis, and oxidative damage-related factors. Western blot determined key proteins in the p38 MAPK signaling. Results indicated that I/R induced the increase of inflammatory factors, lung weight, filtration coefficient, bronchoalveolar lavage fluid protein content, apoptosis, neutrophil, and lung peroxidation, and H/R caused cell growth inhibition, apoptosis, and oxidative damage-related factors' release. EPO attenuated I/R-induced injury in vivo and in vitro. Furthermore, the increase of p-p38, p-JNK, and p-ERK1/2 in lung tissues and cells induced by I/R was downregulated by EPO. Moreover, both EPO and an inhibitor of p38 MAPK (SB203580) alleviated H/R-induced cell injury. Erythropoietin along with SB203580 had more obvious protection effects than EPO alone. Collectively, EPO alleviated I/R-induced ALI by blocking p38 MAPK signaling. The interaction mechanism of EPO with p38 MAPK signaling contributes to understanding the processes of I/R-induced ALI and provides new insights for the disease treatment.
Collapse
Affiliation(s)
- Ling Jia
- Department of Critical Care Medicine, 572527Sir Run Run Hospital Nanjing Medical University, Nanjing, China
| | - Wenjing Cui
- Department of Radiology, 375808Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiao Chen
- Department of Critical Care Medicine, 572527Sir Run Run Hospital Nanjing Medical University, Nanjing, China
| | - Jinghui Yang
- Department of Critical Care Medicine, 572527Sir Run Run Hospital Nanjing Medical University, Nanjing, China
| | - Xiang Xue
- Department of Critical Care Medicine, 572527Sir Run Run Hospital Nanjing Medical University, Nanjing, China
| | - Jianqin Cai
- Department of Critical Care Medicine, 572527Sir Run Run Hospital Nanjing Medical University, Nanjing, China
| | - Wei Zhao
- Department of Critical Care Medicine, 572527Sir Run Run Hospital Nanjing Medical University, Nanjing, China
| | - Wei Gao
- Department of Geriatrics, 572527Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
135
|
Jain A. High circulating Plasma Soluble Receptor for Advanced Glycation End-Products in Early CARDS - Pathophysiological Significance? Am J Respir Crit Care Med 2021; 205:254-256. [PMID: 34727514 PMCID: PMC8787241 DOI: 10.1164/rccm.202108-1896le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Amit Jain
- Cleveland Clinic Abu Dhabi, 284697, Anesthesiology Institute , Abu Dhabi, United Arab Emirates.,Cleveland Clinic Foundation, 2569, Cleveland, Ohio, United States;
| |
Collapse
|
136
|
Saied EM, El-Maradny YA, Osman AA, Darwish AMG, Abo Nahas HH, Niedbała G, Piekutowska M, Abdel-Rahman MA, Balbool BA, Abdel-Azeem AM. A Comprehensive Review about the Molecular Structure of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2): Insights into Natural Products against COVID-19. Pharmaceutics 2021; 13:1759. [PMID: 34834174 PMCID: PMC8624722 DOI: 10.3390/pharmaceutics13111759] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/06/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
In 2019, the world suffered from the emergence of COVID-19 infection, one of the most difficult pandemics in recent history. Millions of confirmed deaths from this pandemic have been reported worldwide. This disaster was caused by SARS-CoV-2, which is the last discovered member of the family of Coronaviridae. Various studies have shown that natural compounds have effective antiviral properties against coronaviruses by inhibiting multiple viral targets, including spike proteins and viral enzymes. This review presents the classification and a detailed explanation of the SARS-CoV-2 molecular characteristics and structure-function relationships. We present all currently available crystal structures of different SARS-CoV-2 proteins and emphasized on the crystal structure of different virus proteins and the binding modes of their ligands. This review also discusses the various therapeutic approaches for COVID-19 treatment and available vaccinations. In addition, we highlight and compare the existing data about natural compounds extracted from algae, fungi, plants, and scorpion venom that were used as antiviral agents against SARS-CoV-2 infection. Moreover, we discuss the repurposing of select approved therapeutic agents that have been used in the treatment of other viruses.
Collapse
Affiliation(s)
- Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
| | - Yousra A. El-Maradny
- Microbiology Department, High Institute of Public Health, Alexandria University, Alexandria 21526, Egypt;
| | - Alaa A. Osman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, New Giza University, Newgiza, km 22 Cairo-Alexandria Desert Road, Cairo 12256, Egypt;
| | - Amira M. G. Darwish
- Food Technology Department, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA City), Alexandria 21934, Egypt;
| | - Hebatallah H. Abo Nahas
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt; (H.H.A.N.); (M.A.A.-R.)
| | - Gniewko Niedbała
- Department of Biosystems Engineering, Faculty of Environmental and Mechanical Engineering, Poznań University of Life Sciences, Wojska Polskiego 50, 60-627 Poznań, Poland;
| | - Magdalena Piekutowska
- Department of Geoecology and Geoinformation, Institute of Biology and Earth Sciences, Pomeranian University in Słupsk, Partyzantów 27, 76-200 Słupsk, Poland;
| | - Mohamed A. Abdel-Rahman
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt; (H.H.A.N.); (M.A.A.-R.)
| | - Bassem A. Balbool
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza 12585, Egypt;
| | - Ahmed M. Abdel-Azeem
- Botany and Microbiology Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
137
|
Roy RK, Sharma U, Wasson MK, Jain A, Hassan MI, Prakash H. Macrophage Activation Syndrome and COVID 19: Impact of MAPK Driven Immune-Epigenetic Programming by SARS-Cov-2. Front Immunol 2021; 12:763313. [PMID: 34659270 PMCID: PMC8519338 DOI: 10.3389/fimmu.2021.763313] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/20/2021] [Indexed: 01/08/2023] Open
Affiliation(s)
- Roshan Kumar Roy
- Amity Institute of Virology and Immunology, Amity University, NOIDA, Udham Singh Nagar, India
| | - Uttam Sharma
- Department of Zoology, Central University of Punjab, Bathinda, India
| | - Mishi Kaushal Wasson
- Amity Institute of Virology and Immunology, Amity University, NOIDA, Udham Singh Nagar, India
| | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University, NOIDA, Udham Singh Nagar, India
| |
Collapse
|
138
|
Kleinehr J, Wilden JJ, Boergeling Y, Ludwig S, Hrincius ER. Metabolic Modifications by Common Respiratory Viruses and Their Potential as New Antiviral Targets. Viruses 2021; 13:2068. [PMID: 34696497 PMCID: PMC8540840 DOI: 10.3390/v13102068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/22/2021] [Accepted: 10/09/2021] [Indexed: 12/11/2022] Open
Abstract
Respiratory viruses are known to be the most frequent causative mediators of lung infections in humans, bearing significant impact on the host cell signaling machinery due to their host-dependency for efficient replication. Certain cellular functions are actively induced by respiratory viruses for their own benefit. This includes metabolic pathways such as glycolysis, fatty acid synthesis (FAS) and the tricarboxylic acid (TCA) cycle, among others, which are modified during viral infections. Here, we summarize the current knowledge of metabolic pathway modifications mediated by the acute respiratory viruses respiratory syncytial virus (RSV), rhinovirus (RV), influenza virus (IV), parainfluenza virus (PIV), coronavirus (CoV) and adenovirus (AdV), and highlight potential targets and compounds for therapeutic approaches.
Collapse
Affiliation(s)
- Jens Kleinehr
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
| | - Janine J. Wilden
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
| | - Yvonne Boergeling
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
| | - Stephan Ludwig
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
- Cells in Motion Interfaculty Centre (CiMIC), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
| | - Eike R. Hrincius
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
| |
Collapse
|
139
|
Timalsina D, Pokhrel KP, Bhusal D. Pharmacologic Activities of Plant-Derived Natural Products on Respiratory Diseases and Inflammations. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1636816. [PMID: 34646882 PMCID: PMC8505070 DOI: 10.1155/2021/1636816] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Respiratory inflammation is caused by an air-mediated disease induced by polluted air, smoke, bacteria, and viruses. The COVID-19 pandemic is also a kind of respiratory disease, induced by a virus causing a serious effect on the lungs, bronchioles, and pharynges that results in oxygen deficiency. Extensive research has been conducted to find out the potent natural products that help to prevent, treat, and manage respiratory diseases. Traditionally, wider floras were reported to be used, such as Morus alba, Artemisia indica, Azadirachta indica, Calotropis gigantea, but only some of the potent compounds from some of the plants have been scientifically validated. Plant-derived natural products such as colchicine, zingerone, forsythiaside A, mangiferin, glycyrrhizin, curcumin, and many other compounds are found to have a promising effect on treating and managing respiratory inflammation. In this review, current clinically approved drugs along with the efficacy and side effects have been studied. The study also focuses on the traditional uses of medicinal plants on reducing respiratory complications and their bioactive phytoconstituents. The pharmacological evidence of lowering respiratory complications by plant-derived natural products has been critically studied with detailed mechanism and action. However, the scientific validation of such compounds requires clinical study and evidence on animal and human models to replace modern commercial medicine.
Collapse
Affiliation(s)
- Deepak Timalsina
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal
| | | | - Deepti Bhusal
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal
| |
Collapse
|
140
|
Kumar S, Çalışkan DM, Janowski J, Faist A, Conrad BCG, Lange J, Ludwig S, Brunotte L. Beyond Vaccines: Clinical Status of Prospective COVID-19 Therapeutics. Front Immunol 2021; 12:752227. [PMID: 34659259 PMCID: PMC8519339 DOI: 10.3389/fimmu.2021.752227] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
Since November 2019 the SARS-CoV-2 pandemic has caused nearly 200 million infection and more than 4 million deaths globally (Updated information from the World Health Organization, as on 2nd Aug 2021). Within only one year into the pandemic, several vaccines were designed and reached approval for the immunization of the world population. The remarkable protective effects of the manufactured vaccines are demonstrated in countries with high vaccination rates, such as Israel and UK. However, limited production capacities, poor distribution infrastructures and political hesitations still hamper the availability of vaccines in many countries. In addition, due to the emergency of SARS-CoV-2 variants with immune escape properties towards the vaccines the global numbers of new infections as well as patients developing severe COVID-19, remains high. New studies reported that about 8% of infected individuals develop long term symptoms with strong personal restrictions on private as well as professional level, which contributes to the long socioeconomic problems caused by this pandemic. Until today, emergency use-approved treatment options for COVID-19 are limited to the antiviral Remdesivir, a nucleoside analogue targeting the viral polymerase, the glucocorticosteroide Dexamethasone as well as neutralizing antibodies. The therapeutic benefits of these treatments are under ongoing debate and clinical studies assessing the efficiency of these treatments are still underway. To identify new therapeutic treatments for COVID-19, now and by the post-pandemic era, diverse experimental approaches are under scientific evaluation in companies and scientific research teams all over the world. To accelerate clinical translation of promising candidates, repurposing approaches of known approved drugs are specifically fostered but also novel technologies are being developed and are under investigation. This review summarizes the recent developments from the lab bench as well as the clinical status of emerging therapeutic candidates and discusses possible therapeutic entry points for the treatment strategies with regard to the biology of SARS-CoV-2 and the clinical course of COVID-19.
Collapse
Affiliation(s)
- Sriram Kumar
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
| | - Duygu Merve Çalışkan
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
| | - Josua Janowski
- Institute of Virology, University of Münster, Münster, Germany
- SP BioSciences Graduate Program, University of Münster, Münster, Germany
| | - Aileen Faist
- Institute of Virology, University of Münster, Münster, Germany
- CiM-IMPRS Graduate Program, University of Münster, Münster, Germany
| | | | - Julius Lange
- Institute of Virology, University of Münster, Münster, Germany
| | - Stephan Ludwig
- Institute of Virology, University of Münster, Münster, Germany
- EvoPAD Research Training Group 2220, University of Münster, Münster, Germany
- CiM-IMPRS Graduate Program, University of Münster, Münster, Germany
- Interdisciplinary Centre for Medical Research, University of Münster, Münster, Germany
| | - Linda Brunotte
- Institute of Virology, University of Münster, Münster, Germany
- Interdisciplinary Centre for Medical Research, University of Münster, Münster, Germany
| |
Collapse
|
141
|
Valipour M, Zarghi A, Ebrahimzadeh MA, Irannejad H. Therapeutic potential of chelerythrine as a multi-purpose adjuvant for the treatment of COVID-19. Cell Cycle 2021; 20:2321-2336. [PMID: 34585628 PMCID: PMC8506812 DOI: 10.1080/15384101.2021.1982509] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Multifunctional nature of phytochemicals and their chemical diversity has attracted attention to develop leads originated from nature to fight COVID-19. Pharmacological activities of chelerythrine and its congeners have been studied and reported in the literature. This compound simultaneously has two key therapeutic effects for the treatment of COVID-19, antiviral and anti-inflammatory activities. Chelerythrine can prevent hyper-inflammatory immune response through regulating critical signaling pathways involved in SARS-CoV-2 infection, such as alteration in Nrf2, NF-κB, and p38 MAPK activities. In addition, chelerythrine has a strong protein kinase C-α/-β inhibitory activity suitable for cerebral vasospasm prevention and eryptosis reduction, as well as beneficial effects in suppressing pulmonary inflammation and fibrosis. In terms of antiviral activity, chelerythrine can fight with SARS-CoV-2 through various mechanisms, such as direct-acting mechanism, viral RNA-intercalation, and regulation of host-based antiviral targets. Although chelerythrine is toxic in vitro, the in vivo toxicity is significantly reduced due to its structural conversion to alkanolamine. Its multifunctional action makes chelerythrine a prominent compound for the treatment of COVID-19. Considering precautions related to the toxicity at higher doses, it is expected that this compound is useful in combination with proper antivirals to reduce the severity of COVID-19 symptoms.
Collapse
Affiliation(s)
- Mehdi Valipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Afshin Zarghi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Ebrahimzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Irannejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
142
|
Richardson PG, Palomo M, Kernan NA, Hildebrandt GC, Chao N, Carreras E. The importance of endothelial protection: the emerging role of defibrotide in reversing endothelial injury and its sequelae. Bone Marrow Transplant 2021; 56:2889-2896. [PMID: 34584241 PMCID: PMC8477726 DOI: 10.1038/s41409-021-01383-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 11/09/2022]
Abstract
Hepatic veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS), a potentially life-threatening complication of hematopoietic cell transplantation (HCT), results from prolonged sinusoidal endothelial cell activation and profound endothelial cell damage, with sequelae. Defibrotide, the only drug approved in the United States and Europe for treating VOD/SOS post-HCT, has European Commission orphan drug designation for preventing graft-versus-host disease (GvHD), associated with endothelial dysfunction. This endothelial cell protector and stabilizing agent restores thrombo-fibrinolytic balance and preserves endothelial homeostasis through antithrombotic, fibrinolytic, anti-inflammatory, anti-oxidative, and anti-adhesive activity. Defibrotide also preserves endothelial cell structure by inhibiting heparanase activity. Evidence suggests that downregulating p38 mitogen-activated protein kinase (MAPK) and histone deacetylases (HDACs) is key to defibrotide’s endothelial protective effects; phosphatidylinositol 3-kinase/Akt (PI3K/AKT) potentially links defibrotide interaction with the endothelial cell membrane and downstream effects. Despite defibrotide’s being most extensively studied in VOD/SOS, emerging preclinical and clinical data support defibrotide for treating or preventing other conditions driven by endothelial cell activation, dysfunction, and/or damage, such as GvHD, transplant-associated thrombotic microangiopathy, or chimeric antigen receptor T-cell (CAR-T) therapy-associated neurotoxicity, underpinned by cytokine release syndrome and endotheliitis. Further preclinical and clinical studies will explore defibrotide’s potential utility in a broader range of disorders resulting from endothelial cell activation and dysfunction.
Collapse
Affiliation(s)
- Paul G Richardson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Marta Palomo
- Barcelona Endothelium Team, Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain.,Hematopathology, Department of Pathology, Centre de Diagnostic Biomedic (CDB), Hospital Clinic de Barcelona, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Nancy A Kernan
- Pediatric BMT Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Nelson Chao
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Enric Carreras
- Barcelona Endothelium Team, Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| |
Collapse
|
143
|
Al-Kuraishy HM, Al-Gareeb AI, Faidah H, Alexiou A, Batiha GES. Testosterone in COVID-19: An Adversary Bane or Comrade Boon. Front Cell Infect Microbiol 2021; 11:666987. [PMID: 34568081 PMCID: PMC8455954 DOI: 10.3389/fcimb.2021.666987] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/16/2021] [Indexed: 12/17/2022] Open
Abstract
COVID-19 is a pandemic disease caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), which leads to pulmonary manifestations like acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). In addition, COVID-19 may cause extra-pulmonary manifestation such as testicular injury. Both high and low levels of testosterone could affect the severity of COVID-19. Herein, there is substantial controversy regarding the potential role of testosterone in SARS-CoV-2 infection and COVID-19 severity. Therefore, the present study aimed to review and elucidate the assorted view of preponderance regarding the beneficial and harmful effects of testosterone in COVID-19. A related literature search in PubMed, Scopus, Web of Science, Google Scholar, and Science Direct was done. All published articles related to the role of testosterone and COVID-19 were included in this mini-review. The beneficial effects of testosterone in COVID-19 are through inhibition of pro-inflammatory cytokines, augmentation of anti-inflammatory cytokines, modulation of the immune response, attenuation of oxidative stress, and endothelial dysfunction. However, its harmful effects in COVID-19 are due to augmentation of transmembrane protease serine 2 (TMPRSS2), which is essential for cleaving and activating SARS-CoV-2 spike protein during acute SARS-CoV-2 infection. Most published studies illustrated that low testosterone levels are linked to COVID-19 severity. A low testosterone level in COVID-19 is mainly due to testicular injury, the primary source of testosterone.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Hani Faidah
- Faculty of Medicine, Umm Al Qura University, Mecca, Saudi Arabia
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, Australia.,AFNP Med Austria, Wien, Austria
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
144
|
Liskova A, Koklesova L, Samec M, Abdellatif B, Zhai K, Siddiqui M, Šudomová M, Hassan ST, Kudela E, Biringer K, Giordano FA, Büsselberg D, Golubnitschaja O, Kubatka P. Targeting phytoprotection in the COVID-19-induced lung damage and associated systemic effects-the evidence-based 3PM proposition to mitigate individual risks. EPMA J 2021; 12:325-347. [PMID: 34367380 PMCID: PMC8329620 DOI: 10.1007/s13167-021-00249-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023]
Abstract
The risks related to the COVID-19 are multi-faceted including but by far not restricted to the following: direct health risks by poorly understood effects of COVID-19 infection, overloaded capacities of healthcare units, restricted and slowed down care of patients with non-communicable disorders such as cancer, neurologic and cardiovascular pathologies, among others; social risks-restricted and broken social contacts, isolation, professional disruption, explosion of aggression in the society, violence in the familial environment; mental risks-loneliness, helplessness, defenceless, depressions; and economic risks-slowed down industrial productivity, broken delivery chains, unemployment, bankrupted SMEs, inflation, decreased capacity of the state to perform socially important programs and to support socio-economically weak subgroups in the population. Directly or indirectly, the above listed risks will get reflected in a healthcare occupation and workload which is a tremendous long-term challenge for the healthcare capacity and robustness. The article does not pretend to provide solutions for all kind of health risks. However, it aims to present the scientific evidence of great clinical utility for primary, secondary, and tertiary care to protect affected individuals in a cost-effective manner. To this end, due to pronounced antimicrobial, antioxidant, anti-inflammatory, and antiviral properties, naturally occurring plant substances are capable to protect affected individuals against COVID-19-associated life-threatening complications such as lung damage. Furthermore, they can be highly effective, if being applied to secondary and tertiary care of noncommunicable diseases under pandemic condition. Thus, the stratification of patients evaluating specific health conditions such as sleep quality, periodontitis, smoking, chronic inflammation and diseases, metabolic disorders and obesity, vascular dysfunction, and cancers would enable effective managemenet of COVID-19-associated complications in primary, secondary, and tertiary care in the context of predictive, preventive, and personalized medicine (3PM).
Collapse
Affiliation(s)
- Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Basma Abdellatif
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, 24144 Qatar
| | - Kevin Zhai
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, 24144 Qatar
| | - Manaal Siddiqui
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, 24144 Qatar
| | - Miroslava Šudomová
- Museum of Literature in Moravia, Klášter 1, 66461, Rajhrad, Czech Republic
| | - Sherif T.S. Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 16500 Prague, Czech Republic
| | - Erik Kudela
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Kamil Biringer
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität, Bonn, Germany
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, 24144 Qatar
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
145
|
Das JK, Roy S, Guzzi PH. Analyzing host-viral interactome of SARS-CoV-2 for identifying vulnerable host proteins during COVID-19 pathogenesis. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 93:104921. [PMID: 34004362 PMCID: PMC8123524 DOI: 10.1016/j.meegid.2021.104921] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023]
Abstract
The development of therapeutic targets for COVID-19 relies on understanding the molecular mechanism of pathogenesis. Identifying genes or proteins involved in the infection mechanism is the key to shedding light on the complex molecular mechanisms. The combined effort of many laboratories distributed throughout the world has produced protein and genetic interactions. We integrated available results and obtained a host protein-protein interaction network composed of 1432 human proteins. Next, we performed network centrality analysis to identify critical proteins in the derived network. Finally, we performed a functional enrichment analysis of central proteins. We observed that the identified proteins are primarily associated with several crucial pathways, including cellular process, signaling transduction, neurodegenerative diseases. We focused on the proteins that are involved in human respiratory tract diseases. We highlighted many potential therapeutic targets, including RBX1, HSPA5, ITCH, RAB7A, RAB5A, RAB8A, PSMC5, CAPZB, CANX, IGF2R, and HSPA1A, which are central and also associated with multiple diseases.
Collapse
Affiliation(s)
- Jayanta Kumar Das
- Department of Pediatrics, School of Medicine, Johns Hopkins University, MD, USA
| | - Swarup Roy
- Network Reconstruction & Analysis (NetRA) Lab, Department of Computer Applications, Sikkim University, Gangtok, India,Corresponding authors
| | - Pietro Hiram Guzzi
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy,Corresponding authors
| |
Collapse
|
146
|
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), has resulted in a pandemic that has had widespread effects on human activities. The clinical presentation of severe COVID-19 includes a broad spectrum of clinical disease, most notably acute respiratory distress syndrome, cytokine release syndrome (CRS), multiorgan failure, and death. Direct viral damage and uncontrolled inflammation have been suggested as contributory factors in COVID-19 disease severity. The COVID-19 pandemic has emphasized the critical role of an effective host immune response in controlling a virus infection and demonstrated the devastating effect of immune dysregulation. Understanding the nature of the immune response to SARS-CoV-2 pathogenesis is key to developing effective treatments for COVID-19. Here, we describe the nature of the dysregulated host immune response in COVID-19, identify potential mechanisms involved in CRS, and discuss potential strategies that can be used to manage immune dysregulation in COVID-19. Expected final online publication date for the Annual Review of Medicine, Volume 73 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA;
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242, USA;
| |
Collapse
|
147
|
Sunkara H, Dewan SMR. Coronavirus disease-2019: A review on the disease exacerbation via cytokine storm and concurrent management. Int Immunopharmacol 2021; 99:108049. [PMID: 34426104 PMCID: PMC8343371 DOI: 10.1016/j.intimp.2021.108049] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
Setting up treatment strategies is the highest concern today to reduce the fatality of COVID-19. Due to a very new kind of virus attack, no specific treatment has been discovered to date. The most crucial way to dominate the disease severity is now the repurposing of drugs. In this review, we focused on the current treatment approaches targeting the crucial causative factors for the disease burden through cytokine storm or cytokine release syndrome. Several vaccines have been developed and have been applied already for prevention purposes, and several are on the way to be developed, although the effects and side effects are under observation. Presently, regulation of the immune response through intervention treatment methods has been adjusted on the basis of the COVID-19 severity stage and generally includes vaccines, immunotherapies including convalescent plasma and immunoglobulin treatment, monoclonal antibodies, cytokine therapy, complement inhibition, regenerative medicine, and repurposed anti-inflammatory and immune-regulatory drugs. Combination therapy is not acceptable in all respects because there is no concrete evidence in clinical trials or in vivo data. Target-specific drug therapies, such as inhibition of cytokine-producing signaling pathways, could be an excellent solution and thus reduce the severity of inflammation and disease severity. Therefore, gathering information about the mechanism of disease progression, possible goals, and drug efficacy of immune-based approaches to combat COVID-19 in the context of orderly review analysis is consequential.
Collapse
Affiliation(s)
- Haripriya Sunkara
- Pharmacology Division, Center for Life Sciences Research, Dhaka, Bangladesh; Department of Pharmacy Practice, Vijaya Institute of Pharmaceutical Sciences for Women, Vijayawada, India
| | - Syed Masudur Rahman Dewan
- Pharmacology Division, Center for Life Sciences Research, Dhaka, Bangladesh; Department of Pharmacy, Noakhali Science and Technology University, Noakhali, Chattogram, Bangladesh; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
148
|
Mozafar M, Mirmotalebisohi SA, Sameni M, Zali H. Investigating Neurological Manifestations of SARS-CoV-2. J Neuroimmune Pharmacol 2021; 16:695-698. [PMID: 34431023 PMCID: PMC8384098 DOI: 10.1007/s11481-021-10006-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/31/2021] [Indexed: 11/01/2022]
Affiliation(s)
- Maryam Mozafar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Seyed Amir Mirmotalebisohi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Sameni
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran. .,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
149
|
Keane C, Coalter M, Martin-Loeches I. Immune System Disequilibrium-Neutrophils, Their Extracellular Traps, and COVID-19-Induced Sepsis. Front Med (Lausanne) 2021; 8:711397. [PMID: 34485339 PMCID: PMC8416266 DOI: 10.3389/fmed.2021.711397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Equilibrium within the immune system can often determine the fate of its host. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) pandemic. Immune dysregulation remains one of the main pathophysiological components of SARS-CoV-2-associated organ injury, with over-activation of the innate immune system, and induced apoptosis of adaptive immune cells. Here, we provide an overview of the innate immune system, both in general and relating to COVID-19. We specifically discuss "NETosis," the process of neutrophil release of their extracellular traps, which may be a more recently described form of cell death that is different from apoptosis, and how this may propagate organ dysfunction in COVID-19. We complete this review by discussing Stem Cell Therapies in COVID-19 and emerging COVID-19 phenotypes, which may allow for more targeted therapy in the future. Finally, we consider the array of potential therapeutic targets in COVID-19, and associated therapeutics.
Collapse
Affiliation(s)
- Colm Keane
- Department of Anaesthesia and Intensive Care, St. James's Hospital, Dublin, Ireland
- Multidisciplinary Intensive Care Research Organization (MICRO), Trinity College Dublin, Dublin, Ireland
| | - Matthew Coalter
- Department of Anaesthesia and Intensive Care, St. James's Hospital, Dublin, Ireland
| | - Ignacio Martin-Loeches
- Department of Anaesthesia and Intensive Care, St. James's Hospital, Dublin, Ireland
- Multidisciplinary Intensive Care Research Organization (MICRO), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
150
|
Budamagunta V, Manohar-Sindhu S, Yang Y, He Y, Traktuev DO, Foster TC, Zhou D. Senescence-associated hyper-activation to inflammatory stimuli in vitro. Aging (Albany NY) 2021; 13:19088-19107. [PMID: 34375950 PMCID: PMC8386536 DOI: 10.18632/aging.203396] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/27/2021] [Indexed: 04/11/2023]
Abstract
Aging is associated with an increased susceptibility to adverse inflammatory conditions such as sepsis and cytokine storm. We hypothesized that senescent cells (SnCs) play a central role in this age-associated pathology in part due to their expression of the senescence-associated secretory phenotype (SASP), which may prime SnCs to inflammatory stimulation. To test this hypothesis, we examined the expression of various inflammatory cytokines and chemokines at the levels of gene transcription and protein production in various SnCs in vitro in response to lipopolysaccharide (LPS), interleukin-1β (IL1β), and tumor necrosis factor α (TNFα) stimulation. We found that SnCs not only expressed higher basal levels of various inflammatory cytokines and chemokines as a manifestation of the SASP, but more importantly exhibited hyper-activation of the induction of a variety of inflammatory mediators in response to LPS, IL1β and TNFα stimulation as compared with non-SnCs. This senescence-associated hyper-activation is likely mediated in part via the p38MAPK (p38) and NFκB pathways because LPS stimulation elicited significantly higher levels of p38 phosphorylation and NFκB p65 nuclear translation in SnCs when compared to their non-senescent counterparts and inhibition of these pathways with losmapimod (a p38 specific inhibitor) and BMS-345541 (a selective NFκB inhibitor) attenuated LPS-induced expression of IL6, TNFα, CCL5, and IL1β mRNA in SnCs. These findings suggest that SnCs may play an important role in the age-related increases in the susceptibility to developing an exacerbated inflammatory response and highlight the potential to use senotherapeutics to ameliorate the severity of various devastating inflammatory conditions in the elderly.
Collapse
Affiliation(s)
- Vivekananda Budamagunta
- Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Sahana Manohar-Sindhu
- Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Yang Yang
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
- Pharmacology and Therapeutics Graduate Program, Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Yonghan He
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Dmitry O. Traktuev
- Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Thomas C. Foster
- Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Daohong Zhou
- Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|