1
|
Al-Naimi MS, Abu-Raghif AR, Fawzi HA. Novel therapeutic effects of rifaximin in combination with methylprednisolone for LPS-induced oxidative stress and inflammation in mice: An in vivo study. Toxicol Rep 2024; 13:101808. [PMID: 39640902 PMCID: PMC11617758 DOI: 10.1016/j.toxrep.2024.101808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 12/07/2024] Open
Abstract
Cytokine-releasing syndrome (CRS) is a special form of systemic inflammatory response syndrome provoked by factors like viral infections and certain immunomodulatory drugs. To elucidate the potential role of rifaximin (RIF) and its combination with methylprednisolone (MP) against the development and progression of CRS in mice. This experiment consists of two parts: protective and therapeutic interventions. The protective experiment: in the induction group, mice received an intraperitoneal injection (IP) of 5 mg/kg lipopolysaccharide (LPS) without intervention. The other group received various drugs before the induction by three days, then observed for an additional two days (50 mg/kg MP, 50 mg/kg RIF, and a combination of 25 mg/kg RIF with 25 mg/kg MP. The second part of the study involves the therapeutic potential; all groups received similar doses of drugs to that received in the prevention groups, except LPS induction was given first, and after one hour, the mice received daily doses of the drugs for five days. At the end of the experiment, blood and tissue samples were obtained. Mice treated with RIF and its combination with MP showed improved serum TNF-α, IL-6, IL-8, IL-1β, INF-γ, MDA, and GSH in both prevention and therapeutic groups. Histopathologically, mice treated with rifaximin and its combination with MP ameliorates the tissue damage in both lung and liver tissues following LPS induction. In conclusion, rifaximin showed protective and therapeutic effects in LPS-induced cytokine storms in mice through anti-inflammatory and antioxidant mechanisms, and its combination with methylprednisolone showed additive/ synergistic action.
Collapse
Affiliation(s)
- Marwa Salih Al-Naimi
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Farahidi University, Baghdad, Iraq
| | - Ahmed R. Abu-Raghif
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | | |
Collapse
|
2
|
Deus MDC, Gadotti AC, Dias ES, Monte Alegre JB, Van Spitzenbergen BAK, Andrade GB, Tozoni SS, Stocco RB, Olandoski M, Tuon FFB, Pinho RA, de Noronha L, Baena CP, Moreno-Amaral AN. Prospective Variation of Cytokine Trends during COVID-19: A Progressive Approach from Disease Onset until Outcome. Int J Mol Sci 2024; 25:10578. [PMID: 39408907 PMCID: PMC11477561 DOI: 10.3390/ijms251910578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/19/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
COVID-19 is characterized by pronounced hypercytokinemia. The cytokine switch, marked by an imbalance between pro-inflammatory and anti-inflammatory cytokines, emerged as a focal point of investigation throughout the COVID-19 pandemic. However, the kinetics and temporal dynamics of cytokine release remain contradictory, making the development of new therapeutics difficult, especially in severe cases. This study collected serum samples from SARS-CoV-2 infected patients at 72 h intervals and monitored them for various cytokines at each timepoint until hospital discharge or death. Cytokine levels were analyzed based on time since symptom onset and patient outcomes. All cytokines studied prospectively were strong predictors of mortality, particularly IL-4 (AUC = 0.98) and IL-1β (AUC = 0.96). First-timepoint evaluations showed elevated cytokine levels in the mortality group (p < 0.001). Interestingly, IFN-γ levels decreased over time in the death group but increased in the survival group. Patients who died exhibited sustained levels of IL-1β and IL-4 and increased IL-6 levels over time. These findings suggest cytokine elevation is crucial in predicting COVID-19 mortality. The dynamic interplay between IFN-γ and IL-4 highlights the balance between Th1/Th2 immune responses and underscores IFN-γ as a powerful indicator of immune dysregulation throughout the infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Andrea Novais Moreno-Amaral
- Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Escola de Medicina, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, PR, Brazil; (M.d.C.D.); (A.C.G.); (E.S.D.); (J.B.M.A.); (B.A.K.V.S.); (G.B.A.); (S.S.T.); (R.B.S.); (M.O.); (F.F.B.T.); (R.A.P.); (L.d.N.); (C.P.B.)
| |
Collapse
|
3
|
Zhao M, Wang B, Zhou F, Fang C, Zhu B, Zhou M, Ye X, Chen Y, Ding Z. Modeling "Two-Hit" Severe Pneumonia in Mice: Pathological Characteristics and Mechanistic Studies. Inflammation 2024:10.1007/s10753-024-02136-w. [PMID: 39212889 DOI: 10.1007/s10753-024-02136-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Severe pneumonia is one of the most common critical diseases in clinical practice. Existing models for severe pneumonia have limitations, leading to limited clinical translation. In this study, a two-hit severe pneumonia mouse model was established by inducing primary pneumonia through intratracheal instillation of 800 μg lipopolysaccharide (LPS), followed by intraperitoneal injection of 10 mg/kg LPS. The effectiveness of various inflammatory indicators and the lung tissue damage during the time course of this model were confirmed and evaluated. At 3 h post two-hit, the IL-6, TNF-α levels in peripheral blood and bronchoalveolar lavage fluid (BALF), and the white blood cells, neutrophils, and lymphocytes in BALF notably exhibited the most pronounced elevation. At 12 h post two-hit, the white blood cells and neutrophils in peripheral blood significantly increased, accompanied by notable alterations in splenic immune cells and worsened pulmonary histopathological damage. Transcriptomics of lung tissue, microbiota analysis of lung and gut, as well as plasma metabolomics analyses further indicated changes in transcriptional profiles, microbial composition, and metabolites due to the two-hit modeling. These results validate that the two-hit model mimics the clinical presentation of severe pneumonia and serves as a robust experimental tool for studying the pathogenesis of severe pneumonia and developing and assessing treatment strategies.
Collapse
Affiliation(s)
- Mengjia Zhao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bixu Wang
- Ningbo Yinzhou Center for Disease Control and Prevention, Ningbo, 315199, China
| | - Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chengnan Fang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bingqi Zhu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mingyuan Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaoqing Ye
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuchi Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
4
|
El-Mezayen NS, Abelrazik YR, Khalifa DM, Dorbouk NM, Moaaz MA, Ali MM, Evy AG, Mohamed EG, Abdelhadi AM, Adly I, Shams NA. Cross-relationship between COVID-19 infection and anti-obesity products efficacy and incidence of side effects: A cross-sectional study. PLoS One 2024; 19:e0309323. [PMID: 39173063 PMCID: PMC11341056 DOI: 10.1371/journal.pone.0309323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/08/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Obesity and COVID-19 are at the top of nowadays health concerns with significant crosstalk between each other. The COVID-19 pandemic negatively affected healthy lifestyles and increased obesity prevalence. Thus, there was a surge in anti-obesity products (AOPs) intake. Herein, we evaluated how the pandemic has affected slimming products' efficacy and safety in patients seeking weight reduction at an urban, weight management centre in Alexandria, Egypt. In addition, the effect of AOPs on COVID-19 infection severity was also appraised to detect whether AOPs can alter COVID-19 host cell entry and infective mechanisms, and thus, affect infection severity. METHODS Patients were invited to complete an anonymous survey. The survey assessed self-reported changes in weight, the use of AOPs during the COVID-19 pandemic, COVID-19 infection severity, AOPs efficacy, and incidence of side effects. Inclusion criteria were obese patients above 18 years old who got infected by COVID-19 while receiving a single-ingredient AOP. RESULTS A total of 462 participants completed our anonymous validated questionnaire. Most of the participants were females (450; 98.4%) with BMI ranging from 24.98-58.46. Eligible participants were only 234 and the top-administered products were orlistat, liraglutide, metformin, green tea, cinnamon, Garcinia cambogia, and Gymnema Sylvestre. In most cases, AOPs intake was beneficial for COVID-19 infection, and most patients experienced mild-to-moderate COVID-19 symptoms. On the other hand, SARS-CoV-2 significantly interferes with AOPs' mechanisms of action which positively or negatively influences their efficacy and side effects incidence due to predictable pharmacological link. CONCLUSION Concurrent AOPs intake with COVID-19 infection is a two-sided weapon; AOPs attenuate COVID-19 infection, while SARS-CoV-2 interferes with efficacy and side effects incidence of AOPs.
Collapse
Affiliation(s)
- Nesrine S. El-Mezayen
- Faculty of Pharmacy, Department of Pharmacology and Therapeutics, Pharos University in Alexandria, Alexandria, Egypt
| | - Yasser R. Abelrazik
- Faculty of Pharmacy, Phftablearos University in Alexandria, Alexandria, Egypt
| | - Dina M. Khalifa
- Faculty of Pharmacy, Phftablearos University in Alexandria, Alexandria, Egypt
| | - Nada M. Dorbouk
- Faculty of Pharmacy, Phftablearos University in Alexandria, Alexandria, Egypt
| | - Mai A. Moaaz
- Faculty of Pharmacy, Phftablearos University in Alexandria, Alexandria, Egypt
| | - Merna M. Ali
- Faculty of Pharmacy, Phftablearos University in Alexandria, Alexandria, Egypt
| | - Alaa G. Evy
- Faculty of Pharmacy, Phftablearos University in Alexandria, Alexandria, Egypt
| | | | - Ahmed M. Abdelhadi
- Faculty of Pharmacy, Phftablearos University in Alexandria, Alexandria, Egypt
| | - Irinie Adly
- Faculty of Pharmacy, Phftablearos University in Alexandria, Alexandria, Egypt
| | | |
Collapse
|
5
|
Nasiri Z, Soleimanjahi H, Baheiraei N, Hashemi SM, Pourkarim MR. The impact understanding of exosome therapy in COVID-19 and preparations for the future approaches in dealing with infectious diseases and inflammation. Sci Rep 2024; 14:5724. [PMID: 38459174 PMCID: PMC10924089 DOI: 10.1038/s41598-024-56334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/05/2024] [Indexed: 03/10/2024] Open
Abstract
Cytokine storms, which result from an abrupt, acute surge in the circulating levels of different pro-inflammatory cytokines, are one of the complications associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This study aimed to assess the effect of exosomes on the release of pro-inflammatory cytokines in patients with coronavirus disease 2019 (COVID-19) and compare it with a control group. The cytokines evaluated in this study were TNF-α, IL-6, IL-17, and IFN-γ. The study compared the levels of these pro-inflammatory cytokines in the peripheral blood mononuclear cells (PBMCs) of five COVID-19 patients in the intensive care unit, who were subjected to both inactivated SARS-CoV-2 and exosome therapy, with those of five healthy controls. The cytokine levels were quantified using the ELISA method. The collected data was analyzed in SPSS Version 26.0 and GraphPad Prism Version 9. According to the study findings, when PBMCs were exposed to inactivated SARS-CoV-2, pro-inflammatory cytokines increased in both patients and healthy controls. Notably, the cytokine levels were significantly elevated in the COVID-19 patients compared to the control group P-values were < 0.001, 0.001, 0.008, and 0.008 for TNF-α, IL-6, IL-17, and IFN-γ, respectively. Conversely, when both groups were exposed to exosomes, there was a marked reduction in the levels of pro-inflammatory cytokines. This suggests that exosome administration can effectively mitigate the hyperinflammation induced by COVID-19 by suppressing the production of pro-inflammatory cytokines in patients. These findings underscore the potential safety and efficacy of exosomes as a therapeutic strategy for COVID-19.
Collapse
Affiliation(s)
- Zeynab Nasiri
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Nafiseh Baheiraei
- Department of Anatomical Science, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Pourkarim
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Leuven, Belgium
| |
Collapse
|
6
|
Wang P, Wang Y, Qin J. Multi-organ microphysiological system: A new paradigm for COVID-19 research. ORGANS-ON-A-CHIP 2023; 5:100029. [PMID: 37206997 PMCID: PMC10181865 DOI: 10.1016/j.ooc.2023.100029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/29/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2, is a systemic disease with a broad spectrum of manifestations in multiple organs. Till now, it remains unclear whether these multi-organ dysfunctions arise from direct viral infection, or indirect injuries. There is an urgent need to evaluate the impacts of SARS-CoV-2 infection on human bodies and explore the pathogenesis of extrapulmonary organ injuries at a systemic level. Multi-organ microphysiological systems, which can model whole-body physiology with engineered tissues and physiological communications between different organs, serve as powerful platforms to model COVID-19 in a multi-organ manner. In this perspective, we summarize the recent advancement in multi-organ microphysiological system-based researches, discuss the remaining challenges, and proposed some prospects in the application of multi-organ model system for COVID-19 research.
Collapse
Affiliation(s)
- Peng Wang
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Yaqing Wang
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Jianhua Qin
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Zivancevic-Simonovic S, Minic R, Cupurdija V, Stanojevic-Pirkovic M, Milosevic-Djordjevic O, Jakovljevic V, Mihaljevic O. Transforming growth factor beta 1 (TGF-β1) in COVID-19 patients: relation to platelets and association with the disease outcome. Mol Cell Biochem 2023; 478:2461-2471. [PMID: 36869188 PMCID: PMC9984293 DOI: 10.1007/s11010-023-04674-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 02/07/2023] [Indexed: 03/05/2023]
Abstract
Transforming growth factor beta (TGF-β) is a ubiquitously distributed cytokine known to contribute to the pathogenesis of numerous pathological processes. The aim of this study was to measure serum concentrations of TGF-β1 in severely ill COVID-19 patients and to analyze its relationship with selected hematological and biochemical parameters and with the disease outcome. The study population included 53 COVID-19 patients with severe clinical expression of the disease and 15 control subjects. TGF-β1 was determined in serum samples and supernatants from PHA-stimulated whole blood cultures using ELISA assay. Biochemical and hematological parameters were analyzed using standard accepted methods. Our results showed that serum levels of TGF-β1 in COVID-19 patients and controls correlate with the platelet counts. Also, positive correlations of TGF-β1 with white blood cell and lymphocyte counts, platelet-to-lymphocyte (PLR) ratio, and fibrinogen level were shown, while negative correlations of this cytokine with platelet distribution width (PDW), D-dimer and activated partial thromboplastin time (a-PTT) values in COVID-19 patients were observed. The lower serum values of TGF-β1 were associated with the unfavorable outcome of COVID-19. In conclusion, TGF-β1 levels were strongly associated with platelet counts and unfavorable disease outcome of severely ill COVID-19 patients.
Collapse
Affiliation(s)
| | - Rajna Minic
- Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Vojislav Cupurdija
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marijana Stanojevic-Pirkovic
- University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Olgica Mihaljevic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
8
|
Pi P, Zeng Z, Zeng L, Han B, Bai X, Xu S. Molecular mechanisms of COVID-19-induced pulmonary fibrosis and epithelial-mesenchymal transition. Front Pharmacol 2023; 14:1218059. [PMID: 37601070 PMCID: PMC10436482 DOI: 10.3389/fphar.2023.1218059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Abstract
As the outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) first broke out in Hubei Province, China, at the end of 2019. It has brought great challenges and harms to global public health. SARS-CoV-2 mainly affects the lungs and is mainly manifested as pulmonary disease. However, one of the biggest crises arises from the emergence of COVID-19-induced fibrosis. At present, there are still many questions about how COVID-19 induced pulmonary fibrosis (PF) occurs and how to treat and regulate its long-term effects. In addition, as an important process of fibrosis, the effect of COVID-19 on epithelial-mesenchymal transition (EMT) may be an important factor driving PF. This review summarizes the main pathogenesis and treatment mechanisms of COVID-19 related to PF. Starting with the basic mechanisms of PF, such as EMT, transforming growth factor-β (TGF-β), fibroblasts and myofibroblasts, inflammation, macrophages, innate lymphoid cells, matrix metalloproteinases and tissue inhibitors of metalloproteinases, hedgehog pathway as well as Notch signaling. Further, we highlight the importance of COVID-19-induced EMT in the process of PF and provide an overview of the related molecular mechanisms, which will facilitate future research to propose new clinical therapeutic solutions for the treatment of COVID-19-induced PF.
Collapse
Affiliation(s)
- Peng Pi
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Zhipeng Zeng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Liqing Zeng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Bing Han
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Xizhe Bai
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Shousheng Xu
- School of Sports Engineering, Beijing Sport University, Beijing, China
| |
Collapse
|
9
|
Li R, Qu S, Qin M, Huang L, Huang Y, Du Y, Yu Z, Fan F, Sun J, Li Q, So KF. Immunomodulatory and antiviral effects of Lycium barbarum glycopeptide on influenza a virus infection. Microb Pathog 2023; 176:106030. [PMID: 36773941 DOI: 10.1016/j.micpath.2023.106030] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Influenza is caused by a respiratory virus and has a major global impact on human health. Influenza A viruses in particular are highly pathogenic to humans and have caused multiple pandemics. An important consequence of infection is viral pneumonia, and with serious complications of excessive inflammation and tissue damage. Therefore, simultaneously reducing direct damage caused by virus infection and relieving indirect damage caused by excessive inflammation would be an effective treatment strategy. Lycium barbarum glycopeptide (LbGp) is a mixture of five highly branched polysaccharide-protein conjuncts (LbGp1-5) isolated from Lycium barbarum fruit. LbGp has pro-immune activity that is 1-2 orders of magnitude stronger than that of other plant polysaccharides. However, there are few reports on the immunomodulatory and antiviral activities of LbGp. In this study, we evaluated the antiviral and immunomodulatory effects of LbGp in vivo and in vitro and investigated its therapeutic effect on H1N1-induced viral pneumonia and mechanisms of action. In vitro, cytokine secretion, NF-κB p65 nuclear translocation, and CD86 mRNA expression in LPS-stimulated RAW264.7 cells were constrained by LbGp treatment. In A549 cells, LbGp can inhibit H1N1 infection by blocking virus attachment and entry action. In vivo experiments confirmed that administration of LbGp can effectively increase the survival rate, body weight and decrease the lung index of mice infected with H1N1. Compared to the model group, pulmonary histopathologic symptoms in lung sections of mice treated with LbGp were obviously alleviated. Further investigation revealed that the mechanism of LbGp in the treatment of H1N1-induced viral pneumonia includes reducing the viral load in lung, regulating the phenotype of pulmonary macrophages, and inhibiting excessive inflammation. In conclusion, LbGp exhibits potential curative effects against H1N1-induced viral pneumonia in mice, and these effects are associated with its good immuno-regulatory and antiviral activities.
Collapse
Affiliation(s)
- Runwei Li
- College of Life Science and Technology, Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No.4 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Shuang Qu
- College of Life Science and Technology, Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Meng Qin
- College of Life Science and Technology, Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lu Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| | - Yichun Huang
- College of Life Science and Technology, Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yi Du
- Center of Clinical Evaluation and Analysis, Pharmacy Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou, 310006, China
| | - Zhexiong Yu
- Ningxia Tianren Goji Biotechnology, Ningxia, 755100, China
| | - Fu Fan
- Ningxia Tianren Goji Biotechnology, Ningxia, 755100, China
| | - Jing Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No.4 Yinghua East Road, Chaoyang District, Beijing, 100029, China.
| | - Qiushuang Li
- Center of Clinical Evaluation and Analysis, Pharmacy Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou, 310006, China.
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
10
|
Silva MJA, Ribeiro LR, Gouveia MIM, Marcelino BDR, dos Santos CS, Lima KVB, Lima LNGC. Hyperinflammatory Response in COVID-19: A Systematic Review. Viruses 2023; 15:553. [PMID: 36851766 PMCID: PMC9962879 DOI: 10.3390/v15020553] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
COVID-19 is a multisystemic disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The immunopathogenic conditions of the hyperinflammatory response that cause systemic inflammation are extremely linked to its severity. This research sought to review the immunopathological elements that contribute to its progression. This is a systematic review using the PUBMED, LILACS, MEDLINE, and SCIELO databases using articles between May 2020 and July 2022 with the following search terms in conjunction with "AND": "SARS-CoV-2"; "COVID-19"; "ARDS" and "Cytokine Storm". The quality appraisal and risk of bias were assessed by the JBI checklists and the Cochrane Collaboration's RoB 2.0 and ROBINS-I tools, respectively, and the risk of bias for in vitro studies by a pre-defined standard in the literature. The search resulted in 39 articles. The main actors in this response denote SARS-CoV-2 Spike proteins, cellular proteases, leukocytes, cytokines, and proteolytic cascades. The "cytokine storm" itself brings several complications to the host through cytokines such as IL-6 and chemokines (such as CCL2), which influence tissue inflammation through apoptosis and pyroptosis. The hyperinflammatory response causes several unfavorable outcomes in patients, and systemic inflammation caused largely by the dysregulation of the immune response should be controlled for their recovery.
Collapse
Affiliation(s)
- Marcos Jessé Abrahão Silva
- Graduate Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
- Bacteriology and Mycology Section, Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
| | - Layana Rufino Ribeiro
- Bacteriology and Mycology Section, Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
| | | | - Beatriz dos Reis Marcelino
- Graduate Program in Parasitic Biology in the Amazon (PPGBPA), University of Pará State (UEPA), Belém 66087-670, PA, Brazil
| | - Carolynne Silva dos Santos
- Federal Institute of Education, Science, and Technology of Pará (IFPA), Abaetetuba 68440-000, PA, Brazil
| | | | - Luana Nepomuceno Gondim Costa Lima
- Graduate Program in Epidemiology and Health Surveillance (PPGEVS), Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
- Bacteriology and Mycology Section, Evandro Chagas Institute (IEC), Ananindeua 67030-000, PA, Brazil
| |
Collapse
|
11
|
Sun Y, Tao Q, Cao Y, Yang T, Zhang L, Luo Y, Wang L. Kaempferol has potential anti-coronavirus disease 2019 (COVID-19) targets based on bioinformatics analyses and pharmacological effects on endotoxin-induced cytokine storm. Phytother Res 2023. [PMID: 36726236 DOI: 10.1002/ptr.7740] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 02/03/2023]
Abstract
COVID-19 has infected 272 million patients and caused 5.33 million deaths around the world, and it remains the main global threat. Previous studies revealed that Chinese traditional medicine is an effective treatment for COVID-19 infection. This study aims to reveal the pharmacological effects of kaempferol, which is the active component of Radix Bupleuri and Tripterygii Radix, and potential mechanisms for the treatment of COVID-19. Here, we employed the bioinformatics methods to filter the anti-COVID-19 candidate genes of kaempferol, which mainly enriched in inflammation (TNF, JUN, etc.) and virus infection (AKT1, JNK, etc.). The Transcription levels of AKT1, JNK and JUN were significantly reduced by kaempferol treatment in the LPS-activated macrophages. In addition, kaempferol reduced the secretion of inflammatory factors by LPS-stimulated macrophages, inhibited MAPK/NF-κB signaling and regulated macrophage polarization to M2 type in vitro, and suppressed endotoxin-induced cytokine storm and improved survival in mice. Molecular docking analysis demonstrated that kaempferol was probable to bind the COVID-19 protein 5R84 and formatted hydrogen bond with the residues, the free binding energy of which was lower than the original ligand. In summary, our current work indicates that kaempferol has anti-COVID-19 potential through the reduction of COVID-19-induced body dysfunction and molecule-protein interaction, and bioinformatics results clarify that some of these key target genes might serve as potential molecular markers for detecting COVID-19.
Collapse
Affiliation(s)
- Yaoxiang Sun
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Qing Tao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yang Cao
- College of Arts & Science, Vanderbilt University, Nashville, Tennessee, USA
| | - Tingting Yang
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Ling Zhang
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Yifeng Luo
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Lei Wang
- Department of Clinical Laboratory, Jiangsu Province hospital on Integration of Chinese and Western Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
12
|
Thabet RH, Massadeh NA, Badarna OB, Al-Momani OM. Highlights on molecular targets in the management of COVID-19: Possible role of pharmacogenomics. J Int Med Res 2023; 51:3000605231153764. [PMID: 36717541 PMCID: PMC9893104 DOI: 10.1177/03000605231153764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
By the end of 2022, there had been a reduction in new cases and deaths caused by coronavirus disease 2019 (COVID-19). At the same time, new variants of the severe acute respiratory syndrome coronavirus 2 virus were being discovered. Critically ill patients with COVID-19 have been found to have high serum levels of proinflammatory cytokines, especially interleukin (IL)-6. COVID-19-related mortality has been attributed in most cases to the cytokine storm caused by increased levels of inflammatory cytokines. Dexamethasone in low doses and immunomodulators such as IL-6 inhibitors are recommended to overcome the cytokine storm. This current narrative review highlights the place of other therapeutic choices such as proteasome inhibitors, protease inhibitors and nuclear factor kappa B inhibitors in the treatment of patients with COVID-19.
Collapse
Affiliation(s)
- Romany H. Thabet
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt,Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan,Romany H. Thabet, Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Shafiq Irshidat Street, Irbid 21163, Jordan.
| | - Noor A. Massadeh
- Internship, Princess Basma Hospital, Ministry of Health, Irbid, Jordan
| | - Omar B. Badarna
- Internship, Princess Basma Hospital, Ministry of Health, Irbid, Jordan
| | - Omar M. Al-Momani
- Internship, Princess Basma Hospital, Ministry of Health, Irbid, Jordan
| |
Collapse
|
13
|
Etemad K, Hatami H, Nikpeyma M, Mowla F, Fallah Atatalab F. Long-term Complications of COVID-19 in Nursing Staff; a Retrospective Longitudinal Study. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2023; 11:e17. [PMID: 36743705 PMCID: PMC9887226 DOI: 10.22037/aaem.v11i1.1785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction Although the lungs are the main target of coronavirus disease (COVID-19), infection is also associated with a high rate of extra-pulmonary complications. This study aimed to evaluate the one-year incidence of long-term COVID-19 complications among the nursing staff. Methods This study is a retrospective longitudinal study. All nursing staff working in public hospitals affiliated with Shahid Beheshti University of Medical Sciences who had been affected with COVID-19 were included in the study via convenience sampling method. The patients were recruited from 20th February 2020 to 20th March 2021. Result 1762 nursing staff with a mean age of 35.08 ± 8.02 (Range: 22-66) years were studied (73.5% female). The results showed that among those who reported at least one type of complication, the duration of complications was significantly longer than 1 week and the median was 8 weeks. The findings demonstrated that complications occurred in 65% of infected females. For men, this rate was 62.3%. Generalized pain, anxiety disorders, and skin complications were among the complications that were significantly more common in women than in men. Additionally, the incidence of anosmia, ageusia, anxiety disorders, and skin complications was significantly higher in younger people than in older people. The comparison of complications revealed that nurses were significantly more likely than other occupations to experience respiratory complications, anosmia and ageusia, generalized pain, and skin complications and that outpatients experienced significantly more complications than hospitalized patients. Conclusion The one-year cumulative incidence of long-term complications in nursing staff with COVID-19 was 64.3%. The most common complications were respiratory complications, generalized pain, anosmia, and ageusia, respectively.
Collapse
Affiliation(s)
- Koorosh Etemad
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti university of medical sciences, Tehran, Iran. ,Safety Promotion and Injury Prevention Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Hatami
- Department of Public Health, School of Public Health and Safety and Environmental and Occupational Hazards Control Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahnaz Nikpeyma
- Hospital infection prevention and control Expert, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Forough Mowla
- Faculty of Nursing, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fallah Atatalab
- School of public health and safety, Shahid Beheshti university of medical sciences, Tehran, Iran. ,Corresponding author: Fatemeh Fallah Atatalab; School of Public Health and Safety, Daneshjoo Blvd., Evin Ave., Tehran, Iran. Postal Code: 1983535511, , Tel: 0098-21-22432040-41
| |
Collapse
|
14
|
Saifi MA, Bansod S, Godugu C. COVID-19 and fibrosis: Mechanisms, clinical relevance, and future perspectives. Drug Discov Today 2022; 27:103345. [PMID: 36075378 PMCID: PMC9444298 DOI: 10.1016/j.drudis.2022.103345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/19/2022] [Accepted: 09/01/2022] [Indexed: 01/08/2023]
Abstract
Coronavirus 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has had significant impacts worldwide since its emergence in December, 2019. Despite a high recovery rate, there is a growing concern over its residual, long-term effects. However, because of a lack of long-term data, we are still far from establishing a consensus on post-COVID-19 complications. The deposition of excessive extracellular matrix (ECM), known as fibrosis, has been observed in numerous survivors of COVID-19. Given the exceptionally high number of individuals affected, there is an urgent need to address the emergence of fibrosis post-COVID-19. In this review, we discuss the clinical relevance of COVID-19-associated fibrosis, the current status of antifibrotic agents, novel antifibrotic targets, and challenges to its management.
Collapse
Affiliation(s)
- Mohd Aslam Saifi
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500 037, India
| | - Sapana Bansod
- Department of Internal Medicine, Oncology Division, Washington University, School of Medicine, St Louis, MO 63110, USA
| | - Chandraiah Godugu
- Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500 037, India.
| |
Collapse
|
15
|
Zare Marzouni H, Rahbar M, Seddighi N, Nabizadeh M, Meidaninikjeh S, Sabouni N. Antibody Therapy for COVID-19: Categories, Pros, and Cons. Viral Immunol 2022; 35:517-528. [PMID: 36201297 DOI: 10.1089/vim.2021.0160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is a life-threatening respiratory disease triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It has been considered a pandemic viral infection since December 2019. The investigation of the effective prophylaxis or therapeutic strategies for emergency management of the current condition has become a priority for medical research centers and pharmaceutical companies. This article provides a comprehensive review of antibody therapy and its different categories with their advantages and disadvantages for COVID-19 over the last few years of the current pandemic. Antibodies can be generated by active immunization, including natural infection with a pathogen and vaccination, or by the passive immunization method such as convalescent plasma therapy (CPT) and antibody synthesis in laboratories. Each of these ways has its characteristics. Arming the immune system with antibodies is the main aim of antiviral therapeutic procedures toward SARS-CoV-2. Collecting and discussing various aspects of available data in this field can give researchers a better perspective for the production of antibody-based products or selection of the most appropriate approach of antibody therapies to improve different cases of COVID-19. Moreover, it can help them control similar viral pandemics that may happen in the future appropriately.
Collapse
Affiliation(s)
- Hadi Zare Marzouni
- Qaen School of Nursing and Midwifery, Birjand University of Medical Sciences, Birjand, Iran
| | - Marjan Rahbar
- Department of Food Science and Technology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nazanin Seddighi
- Qaen School of Nursing and Midwifery, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Nabizadeh
- Department of Biology, Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sepideh Meidaninikjeh
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran.,Cancer Biomedical Center (CBC) Research Institute, Tehran, Iran
| | - Nasim Sabouni
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Sodagar A, Javed R, Tahir H, Razak SIA, Shakir M, Naeem M, Yusof AHA, Sagadevan S, Hazafa A, Uddin J, Khan A, Al-Harrasi A. Pathological Features and Neuroinflammatory Mechanisms of SARS-CoV-2 in the Brain and Potential Therapeutic Approaches. Biomolecules 2022; 12:971. [PMID: 35883527 PMCID: PMC9313047 DOI: 10.3390/biom12070971] [Citation(s) in RCA: 13] [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: 06/13/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
The number of deaths has been increased due to COVID-19 infections and uncertain neurological complications associated with the central nervous system. Post-infections and neurological manifestations in neuronal tissues caused by COVID-19 are still unknown and there is a need to explore how brainstorming promoted congenital impairment, dementia, and Alzheimer's disease. SARS-CoV-2 neuro-invasion studies in vivo are still rare, despite the fact that other beta-coronaviruses have shown similar properties. Neural (olfactory or vagal) and hematogenous (crossing the blood-brain barrier) pathways have been hypothesized in light of new evidence showing the existence of SARS-CoV-2 host cell entry receptors into the specific components of human nerve and vascular tissue. Spike proteins are the primary key and structural component of the COVID-19 that promotes the infection into brain cells. Neurological manifestations and serious neurodegeneration occur through the binding of spike proteins to ACE2 receptor. The emerging evidence reported that, due to the high rate in the immediate wake of viral infection, the olfactory bulb, thalamus, and brain stem are intensely infected through a trans-synaptic transfer of the virus. It also instructs the release of chemokines, cytokines, and inflammatory signals immensely to the blood-brain barrier and infects the astrocytes, which causes neuroinflammation and neuron death; and this induction of excessive inflammation and immune response developed in more neurodegeneration complications. The present review revealed the pathophysiological effects, molecular, and cellular mechanisms of possible entry routes into the brain, pathogenicity of autoantibodies and emerging immunotherapies against COVID-19.
Collapse
Affiliation(s)
- Aisha Sodagar
- Department of Botany, Faculty of Sciences, University of Agriculture, Faisalabad 38040, Pakistan;
| | - Rasab Javed
- Institute of Microbiology, University of Agriculture, Faisalabad 38040, Pakistan;
| | - Hira Tahir
- Department of Botany, Government College Women University Faisalabad, Faisalabad 38000, Pakistan;
| | - Saiful Izwan Abd Razak
- Bioinspired Device and Tissue Engineering Research Group, School of Biomedical Engineering and Health Sciences, Faculty of Engineering, Universiti Teknologi Malaysia, Johor Bahru 81310, Johor, Malaysia;
- Sports Innovation & Technology Centre, Institute of Human Centred Engineering, Universiti Teknologi Malaysia, Johor Bahru 81310, Johor, Malaysia
| | - Muhammad Shakir
- School of Life Sciences, Northeast Normal University, Changchun 130024, China;
| | - Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang 050024, China;
| | - Abdul Halim Abdul Yusof
- School of Chemical and Energy Engineering, Faculty of Engineering, Universiti Teknologi Malaysia, Johor Bahru 81310, Johor, Malaysia;
| | - Suresh Sagadevan
- Nanotechnology & Catalysis Research Centre, University of Malaya, Kuala Lumpur 50603, Kuala Lumpur, Malaysia;
| | - Abu Hazafa
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad 38040, Pakistan
| | - Jalal Uddin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, Nizwa 616, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, Nizwa 616, Oman
| |
Collapse
|
17
|
Protective effects of monoammonium glycyrrhizinate on fatty deposit degeneration induced in primary calf hepatocytes by sodium oleate administration in vitro. Res Vet Sci 2022; 150:213-223. [DOI: 10.1016/j.rvsc.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 04/20/2022] [Accepted: 05/04/2022] [Indexed: 11/20/2022]
|
18
|
Yuan H, Liu L, Zhou J, Zhang T, Daily JW, Park S. Bioactive Components of Houttuynia cordata Thunb and Their Potential Mechanisms Against COVID-19 Using Network Pharmacology and Molecular Docking Approaches. J Med Food 2022; 25:355-366. [PMID: 35438554 DOI: 10.1089/jmf.2021.k.0144] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We investigated the molecular mechanism by which Houttuynia cordata Thunb (HCT) may intervene in coronavirus disease 2019 (COVID-19) and COVID-19-induced cytokine storms using network pharmacology and molecular docking approaches. Using the Traditional Chinese medicine Systems Pharmacology Database and Analysis Platform (TCMSP), a "component-target-pathway" topology map of HCT for COVID-19 treatment was constructed using Cytoscape. Core target genes were analyzed using the STRING database, and the signal pathway map and biological mechanism of COVID-19 therapy were obtained using cluster profilers. Active components of HCT were docked with severe respiratory syndrome coronavirus 2 (SARS-CoV-2) 3C-like protease (3CLpro) and RNA-dependent RNA polymerase (RdRp) using AutoDockTools. Data visualization and statistical analysis were conducted using the R program. A molecular dynamic simulation was carried out with the Groningen Machine for Chemical Simulation program. HCT had six active anti-COVID-19 ingredients and 45 molecular targets. Their crucial target proteins for COVID-19 treatment were the RELA (nuclear factor kappa B [NF-κB] p65 subunit), interleukin 6, and mitogen-activated protein kinase 1. In functional enrichment analysis, the potential molecular targets of active components of HCT for COVID-19 treatment belonged to 18 signaling pathways (adjusted P = 2.12E-11). Gene ontology obtained by Kyoto Encyclopedia of Genes and Genome enrichment screening showed that the primary mechanism of COVID-19 treatment was upregulation of protein kinase C followed by downregulations of T cell differentiation and proliferation and NF-κB signaling. Molecular docking showed that the active components of HCT (quercetin and kaempferol) had similar binding affinities for SARS-CoV-2 3CLpro and SARS-CoV-2 RdRp, primary COVID-19 target proteins as did clinically used drugs. These results were confirmed with molecular dynamics simulation. In conclusion, multiple components of HCT, especially quercetin and kaempferol, have the potential to treat COVID-19 infection and COVID-19-induced cytokine storm by targeting multiple proteins.
Collapse
Affiliation(s)
- Heng Yuan
- Department of Bio-Convergence System, Hoseo University, Asan, South Korea
| | - Liping Liu
- College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, China
| | - Junyu Zhou
- Department of Bio-Convergence System, Hoseo University, Asan, South Korea
| | - Ting Zhang
- Department of Bio-Convergence System, Hoseo University, Asan, South Korea
| | - James W Daily
- Daily Manufacturing, Inc., Rockwell, North Carolina, USA
| | - Sunmin Park
- Department of Bio-Convergence System, Hoseo University, Asan, South Korea.,Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan, South Korea
| |
Collapse
|
19
|
Khorasanchi Z, Jafazadeh Esfehani A, Sharifan P, Hasanzadeh E, Shadmand Foumani Moghadam MR, Ahmadi O, Ebrahimi R, Lotfi SZ, Milani N, Mozdourian M, Rezvani R, Vatanparast H, Assaran Darban R, Ferns G, Ghayour Mobarhan M. The effects of high dose vitamin D supplementation as a nutritional intervention strategy on biochemical and inflammatory factors in adults with COVID-19: Study protocol for a randomized controlled trial. Nutr Health 2022; 28:311-317. [PMID: 35322711 PMCID: PMC8948538 DOI: 10.1177/02601060221082384] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Introduction: Low serum vitamin D has been shown to be a risk factor
for Coronavirus 2019 (COVID-19). The aim of this study was to assess the effects
of high dose vitamin D supplementation on hs-CRP, ESR and clinical outcomes,
including duration of hospitalization, quality of life and New York Heart
Association (NYHA) Functional Classification, in adults with COVID-19.
Methods: This double-blind, randomized control trial will be
conducted on patients with RT-PCR and/or chest CT scan diagnosis of COVID-19
admitted in Imam Reza Hospital, Mashhad, Iran. Participants will be randomized
into control and intervention groups based on randomization sampling. The
intervention group will receive soft gel containing 50,000 IU vitamin D on the
first day followed by 10,000 IU/day through a supplement drop daily for 29 days.
The control group will receive 1000 IU vitamin D daily through supplement drop
and a placebo soft gel. All participants will undergo laboratory assessment
including inflammatory markers, serum 25)OH)D, complete blood count (CBC), liver
and renal profile, lipid profile and erythrocyte sedimentation rate (ESR) at
baseline and at day 30. The mortality rate will be recorded in both groups.
Results: Data will be presented using descriptive statistics.
Comparison of changes in study parameters over the study period will be
performed using analysis of covariance adjusting for possible confounders.
Conclusions: The findings of this will provide evidence on the
effects of high dose vitamin D supplementation on inflammatory markers in
hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Zahra Khorasanchi
- Department of Nutrition, School of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Jafazadeh Esfehani
- Metabolic Syndrome Research Center, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Payam Sharifan
- Department of Nutrition, School of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elahe Hasanzadeh
- International UNESCO center for Health Related Basic Sciences and Human Nutrition, Department of Nutrition, Faculty of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Omid Ahmadi
- Department of Parasitology and Mycology, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| | - Reyhaneh Ebrahimi
- International UNESCO center for Health Related Basic Sciences and Human Nutrition, Department of Nutrition, Faculty of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyede Zahra Lotfi
- Kidney Transplantation Complication Research Center, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nasrin Milani
- Department of Internal Medicine, Faculty of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahnaz Mozdourian
- Lung Diseases Research Center, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Rezvani
- Department of Nutrition, School of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hasan Vatanparast
- College of Pharmacy and Nutrition, 7235University of Saskatchewan, Health Sciences E-Wing, Saskatoon, Saskatchewan, Canada
| | - Reza Assaran Darban
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Gordon Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Brighton, UK
| | - Majid Ghayour Mobarhan
- International UNESCO center for Health Related Basic Sciences and Human Nutrition, Department of Nutrition, Faculty of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Cytokine producing ability of peripheral blood cells from COVID-19 patients after unspecific in vitro stimulation. Inflamm Res 2022; 71:331-341. [PMID: 35157090 PMCID: PMC8853300 DOI: 10.1007/s00011-022-01543-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/29/2021] [Accepted: 01/29/2022] [Indexed: 11/05/2022] Open
Abstract
Objective and design Subjects and methods Results Conclusion
Collapse
|
21
|
Zinellu A, Paliogiannis P, Carru C, Mangoni AA. Serum hydroxybutyrate dehydrogenase and COVID-19 severity and mortality: a systematic review and meta-analysis with meta-regression. Clin Exp Med 2021; 22:499-508. [PMID: 34799779 PMCID: PMC8603904 DOI: 10.1007/s10238-021-00777-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/06/2021] [Indexed: 12/14/2022]
Abstract
Alterations in cardiac and renal biomarkers have been reported in coronavirus disease 19 (COVID-19). We conducted a systematic review and meta-analysis to investigate serum concentrations of hydroxybutyrate dehydrogenase (HBDH), a combined marker of myocardial and renal injury, in hospitalized COVID-19 patients with different disease severity and survival status. We searched PubMed, Web of Science and Scopus, between December 2019 and April 2021, for studies reporting HBDH in COVID-19. Risk of bias was assessed using the Newcastle–Ottawa scale, publication bias was assessed with the Begg’s and Egger’s tests, and certainty of evidence was assessed using GRADE. In 22 studies in 15,019 COVID-19 patients, serum HBDH concentrations on admission were significantly higher in patients with high disease severity or non-survivor status when compared to patients with low severity or survivor status (standardized mean difference, SMD = 0.90, 95% CI 0.74 to 1.07, p < 0.001; moderate certainty of evidence). Extreme between-study heterogeneity was observed (I2 = 93.5%, p < 0.001). Sensitivity analysis, performed by sequentially removing each study and re-assessing the pooled estimates, showed that the magnitude and the direction of the effect size were not substantially modified. A significant publication bias was observed. In meta-regression, the SMD of HBDH concentrations was significantly associated with markers of inflammation, sepsis, liver damage, non-specific tissue damage, myocardial injury, and renal function. Higher HBDH concentrations were significantly associated with higher COVID-19 severity and mortality. This biomarker of cardiac and renal injury might be useful for risk stratification in COVID-19. (PROSPERO registration number: CRD42021258123).
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, 5042, Australia.
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
| |
Collapse
|
22
|
Olajide OA, Iwuanyanwu VU, Adegbola OD, Al-Hindawi AA. SARS-CoV-2 Spike Glycoprotein S1 Induces Neuroinflammation in BV-2 Microglia. Mol Neurobiol 2021; 59:445-458. [PMID: 34709564 PMCID: PMC8551352 DOI: 10.1007/s12035-021-02593-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023]
Abstract
In addition to respiratory complications produced by SARS‐CoV‐2, accumulating evidence suggests that some neurological symptoms are associated with the disease caused by this coronavirus. In this study, we investigated the effects of the SARS‐CoV‐2 spike protein S1 stimulation on neuroinflammation in BV-2 microglia. Analyses of culture supernatants revealed an increase in the production of TNF-α, IL-6, IL-1β and iNOS/NO. S1 also increased protein levels of phospho-p65 and phospho-IκBα, as well as enhanced DNA binding and transcriptional activity of NF-κB. These effects of the protein were blocked in the presence of BAY11-7082 (1 µM). Exposure of S1 to BV-2 microglia also increased the protein levels of NLRP3 inflammasome and enhanced caspase-1 activity. Increased protein levels of p38 MAPK was observed in BV-2 microglia stimulated with the spike protein S1 (100 ng/ml), an action that was reduced in the presence of SKF 86,002 (1 µM). Results of immunofluorescence microscopy showed an increase in TLR4 protein expression in S1-stimulated BV-2 microglia. Furthermore, pharmacological inhibition with TAK 242 (1 µM) and transfection with TLR4 small interfering RNA resulted in significant reduction in TNF-α and IL-6 production in S1-stimulated BV-2 microglia. These results have provided the first evidence demonstrating S1-induced neuroinflammation in BV-2 microglia. We propose that induction of neuroinflammation by this protein in the microglia is mediated through activation of NF-κB and p38 MAPK, possibly as a result of TLR4 activation. These results contribute to our understanding of some of the mechanisms involved in CNS pathologies of SARS-CoV-2.
Collapse
Affiliation(s)
- Olumayokun A Olajide
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK.
| | - Victoria U Iwuanyanwu
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Oyinkansola D Adegbola
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Alaa A Al-Hindawi
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| |
Collapse
|
23
|
Olajide OA, Iwuanyanwu VU, Lepiarz-Raba I, Al-Hindawi AA, Aderogba MA, Sharp HL, Nash RJ. Garcinia kola and garcinoic acid suppress SARS-CoV-2 spike glycoprotein S1-induced hyper-inflammation in human PBMCs through inhibition of NF-κB activation. Phytother Res 2021; 35:6963-6973. [PMID: 34697842 PMCID: PMC8661957 DOI: 10.1002/ptr.7315] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/14/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Symptoms and complications associated with severe SARS-CoV-2 infection such as acute respiratory distress syndrome (ARDS) and organ damage have been linked to SARS-CoV-2 spike protein S1-induced increased production of pro-inflammatory cytokines by immune cells. In this study, the effects of an extract of Garcinia kola seeds and garcinoic acid were investigated in SARS-CoV-2 spike protein S1-stimulated human PBMCs. Results of ELISA experiments revealed that Garcinia kola extract (6.25, 12.5, and 25 μg/ml) and garcinoic acid (1.25, 2.5, and 5 μM) significantly reduced SARS-CoV-2 spike protein S1-induced secretion of TNFα, IL-6, IL-1β, and IL-8 in PBMCs. In-cell western assays showed that pre-treatment with Garcinia kola extract and garcinoic acid reduced expressions of both phospho-p65 and phospho-IκBα proteins, as well as NF-κB DNA binding capacity and NF-κB-driven luciferase expression following stimulation of PBMCs with spike protein S1. Furthermore, pre-treatment of PBMCs with Garcinia kola extract prior to stimulation with SARS-CoV-2 spike protein S1 resulted in reduced damage to adjacent A549 lung epithelial cells. These results suggest that the seed of Garcinia kola and garcinoic acid are natural products which may possess pharmacological/therapeutic benefits in reducing cytokine storm in severe SARS-CoV-2 and other coronavirus infections.
Collapse
Affiliation(s)
- Olumayokun A Olajide
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Victoria U Iwuanyanwu
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Izabela Lepiarz-Raba
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Alaa A Al-Hindawi
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Mutalib A Aderogba
- Department of Chemistry, Faculty of Science, Obafemi Awolowo University, Ile-Ife, Nigeria
| | | | | |
Collapse
|
24
|
Singh E, Matada GSP, Abbas N, Dhiwar PS, Ghara A, Das A. Management of COVID-19-induced cytokine storm by Keap1-Nrf2 system: a review. Inflammopharmacology 2021; 29:1347-1355. [PMID: 34373972 PMCID: PMC8352144 DOI: 10.1007/s10787-021-00860-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022]
Abstract
The natural pathway of antioxidant production is mediated through Kelch-like erythroid cell-derived protein with Cap and collar homology [ECH]-associated protein 1 (Keap1)-Nuclear factor erythroid 2-related factor 2 (Nrf2) system. Keap1 maintains a low level of Nrf2 by holding it in its protein complex. Also, Keap1 facilitates the degradation of Nrf2 by ubiquitination. In other words, Keap1 is a down-regulator of Nrf2. To boost the production of biological antioxidants, Keap1 has to be inhibited and Nrf2 has to be released. Liberated Nrf2 is in an unbound state, so it travels to the nucleus to stimulate the antioxidant response element (ARE) present on the antioxidant genes. AREs activate biosynthesis of biological antioxidants through genes responsible for the production of antioxidants. In some cases of coronavirus disease 2019 (COVID-19), there is an enormous release of cytokines. The antioxidant defense mechanism in the body helps in counteracting symptoms induced by the cytokine storm in COVID-19. So, boosting the production of antioxidants is highly desirable in such a condition. In this review article, we have compiled the role of Keap1-Nrf2 system in antioxidant production. We further propose its potential therapeutic use in managing cytokine storm in COVID-19.
Collapse
Affiliation(s)
- Ekta Singh
- Department of Pharmaceutical Chemistry, Acharya and BM Reddy College of Pharmacy, Bengaluru, Karnataka, 560107, India
| | | | - Nahid Abbas
- Department of Pharmaceutical Chemistry, Acharya and BM Reddy College of Pharmacy, Bengaluru, Karnataka, 560107, India
| | - Prasad Sanjay Dhiwar
- Department of Pharmaceutical Chemistry, Acharya and BM Reddy College of Pharmacy, Bengaluru, Karnataka, 560107, India
| | - Abhishek Ghara
- Department of Pharmaceutical Chemistry, Acharya and BM Reddy College of Pharmacy, Bengaluru, Karnataka, 560107, India
| | - Arka Das
- Department of Pharmaceutical Chemistry, Acharya and BM Reddy College of Pharmacy, Bengaluru, Karnataka, 560107, India
| |
Collapse
|
25
|
Shawki MA, Elsayed NS, Mantawy EM, Said RS. Promising drug repurposing approach targeted for cytokine storm implicated in SARS-CoV-2 complications. Immunopharmacol Immunotoxicol 2021; 43:395-409. [PMID: 34057871 PMCID: PMC8171013 DOI: 10.1080/08923973.2021.1931302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/04/2021] [Indexed: 12/16/2022]
Abstract
A global threat has emerged in 2019 due to the rapid spread of Coronavirus disease (COVID-19). As of January 2021, the number of cases worldwide reached 103 million cases and 2.22 million deaths which were confirmed as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This global pandemic galvanized the scientific community to study the causative virus (SARS-CoV2) pathogenesis, transmission, and clinical symptoms. Remarkably, the most common complication associated with this disease is the cytokine storm which is responsible for COVID-19 mortality. Thus, targeting the cytokine storm with new medications is needed to hamper COVID-19 complications where the most prominent strategy for the treatment is drug repurposing. Through this strategy, several steps are skipped especially those required for testing drug safety and thus may help in reducing the dissemination of this pandemic. Accordingly, the aim of this review is to outline the pathogenesis, clinical features, and immune complications of SARS-CoV2 in addition to suggesting several repurposed drugs with their plausible mechanism of action for possible management of severe COVID-19 cases.
Collapse
Affiliation(s)
- May Ahmed Shawki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Noha Salah Elsayed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M. Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Riham S. Said
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
26
|
da Cunha LNOL, Tizziani T, Souza GB, Moreira MA, Neto JSS, Dos Santos CVD, de Carvalho MG, Dalmarco EM, Turqueti LB, Scotti MT, Scotti L, de Assis FF, Braga A, Sandjo LP. Natural Products with tandem Anti-inflammatory, Immunomodulatory and Anti-SARS-CoV/2 effects: A Drug Discovery Perspective against SARS-CoV-2. Curr Med Chem 2021; 29:2530-2564. [PMID: 34313197 DOI: 10.2174/0929867328666210726094955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND COVID-19 is still causing victims with long-term health consequences, mass deaths, and collapsing healthcare systems around the world. The disease has no efficient drugs. However, previous studies revealed that SARS-CoV-2 and SARS-CoV have 96% and 86.5% similarities in cysteine proteases (3CLpro) and papain-like protease (PLpro) sequences, respectively. This resemblance could be significant in the search for drug candidates with antiviral effects against SARS-CoV-2. OBJECTIVE This paper is a compilation of natural products that inhibit SARS-CoV 3CLpro and PLpro and, concomitantly, reduce inflammation and/or modulate the immune system as a perspective strategy for COVID-19 drug discovery. It also presents in silico studies performed on these selected natural products using SARS-CoV-2 3CLpro and PLpro as targets to propose a list of hit compounds. METHOD The plant metabolites were selected in the literature based on their biological activities on SARS-CoV proteins, inflammatory mediators, and immune response. The consensus docking analysis was performed using four different packages. RESULTS Seventy-nine compounds reported in the literature with inhibitory effects on SARS-CoV proteins were reported as anti-inflammatory agents. Fourteen of them showed in previous studies immunomodulatory effects. Five and six of these compounds showed significant in silico consensus as drug candidates that can inhibit PLpro and 3CLpro, respectively. Our findings corroborated recent results reported on anti-SARS-CoV-2 in the literature. CONCLUSION This study revealed that amentoflavone, rubranoside B, savinin, psoralidin, hirsutenone, and papyriflavonol A are good drug candidate for the search of antibiotics against COVID-19.
Collapse
Affiliation(s)
- Luana N O Leal da Cunha
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Tiago Tizziani
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Gabriella B Souza
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Monalisa A Moreira
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - José S S Neto
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Carlos V D Dos Santos
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Maryelle G de Carvalho
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Eduardo M Dalmarco
- Programa de Pós-Graduação em Farmácia, CCS, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Leonardo B Turqueti
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Marcus Tullius Scotti
- Program in Natural and Synthetic Bioactive Products Federal University of Paraíba Cidade Universitária-Castelo Branco III, João Pessoa, PB, Brazil
| | - Luciana Scotti
- Program in Natural and Synthetic Bioactive Products Federal University of Paraíba Cidade Universitária-Castelo Branco III, João Pessoa, PB, Brazil
| | - Francisco F de Assis
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Antonio Braga
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Louis Pergaud Sandjo
- Programa de Pós-Graduação em Química, CFM, Department of Chemistry, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
27
|
Lai A, Chang ML, O'Donnell RP, Zhou C, Sumner JA, Hsiai TK. Association of COVID-19 transmission with high levels of ambient pollutants: Initiation and impact of the inflammatory response on cardiopulmonary disease. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 779:146464. [PMID: 33961545 PMCID: PMC7960028 DOI: 10.1016/j.scitotenv.2021.146464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 05/14/2023]
Abstract
Ambient air pollution contributes to 7 million premature deaths annually. Concurrently, the ongoing coronavirus disease 2019 (COVID-19) pandemic, complicated with S-protein mutations and other variants, caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in over 2.5 million deaths globally. Chronic air pollution-mediated cardiopulmonary diseases have been associated with an increased incidence of hospitalization and mechanical ventilation following COVID-19 transmission. While the underlying mechanisms responsible for this association remain elusive, air pollutant-induced vascular oxidative stress and inflammatory responses have been implicated in amplifying COVID-19-mediated cytokine release and vascular thrombosis. In addition, prolonged exposure to certain types of particulate matter (PM2.5, d < 2.5 μm) has also been correlated with increased lung epithelial and vascular endothelial expression of the angiotensin-converting enzyme-2 (ACE2) receptors to which the SARS-CoV-2 spike glycoproteins (S) bind for fusion and internalization into host cells. Emerging literature has linked high rates of SARS-CoV-2 infection to regions with elevated levels of PM2.5, suggesting that COVID-19 lockdowns have been implicated in regional reductions in air pollutant-mediated cardiopulmonary effects. Taken together, an increased incidence of SARS-CoV-2-mediated cardiopulmonary diseases seems to overlap with highly polluted regions. To this end, we will review the redox-active components of air pollutants, the pathophysiology of SARS-CoV-2 transmission, and the key oxidative mechanisms and ACE2 overexpression underlying air pollution-exacerbated SARS-CoV-2 transmission.
Collapse
Affiliation(s)
- Angela Lai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America
| | - Megan L Chang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America
| | - Ryan P O'Donnell
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Jennifer A Sumner
- Department of Psychology, College of Life Sciences, University of California, Los Angeles, United States of America
| | - Tzung K Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America; Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA, United States of America; Department of Bioengineering, Henry Samueli School of Engineering & Applied Science, University of California, Los Angeles, CA, United States of America.
| |
Collapse
|
28
|
Induction of Exaggerated Cytokine Production in Human Peripheral Blood Mononuclear Cells by a Recombinant SARS-CoV-2 Spike Glycoprotein S1 and Its Inhibition by Dexamethasone. Inflammation 2021; 44:1865-1877. [PMID: 33860869 PMCID: PMC8050229 DOI: 10.1007/s10753-021-01464-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/30/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023]
Abstract
An understanding of the pathological inflammatory mechanisms involved in SARS-CoV-2 virus infection is necessary in order to discover new molecular pharmacological targets for SARS-CoV-2 cytokine storm. In this study, the effects of a recombinant SARS-CoV-2 spike glycoprotein S1 was investigated in human peripheral blood mononuclear cells (PBMCs). Stimulation of PBMCs with spike glycoprotein S1 (100 ng/mL) resulted in significant elevation in the production of TNFα, IL-6, IL-1β and IL-8. However, pre-treatment with dexamethasone (100 nM) caused significant reduction in the release of these cytokines. Further experiments revealed that S1 stimulation of PBMCs increased phosphorylation of NF-κB p65 and IκBα, and IκBα degradation. DNA binding of NF-κB p65 was also significantly increased following stimulation with spike glycoprotein S1. Treatment of PBMCs with dexamethasone (100 nM) or BAY11-7082 (1 μM) resulted in inhibition of spike glycoprotein S1-induced NF-κB activation. Activation of p38 MAPK by S1 was blocked in the presence of dexamethasone and SKF 86002. CRID3, but not dexamethasone pre-treatment, produced significant inhibition of S1-induced activation of NLRP3/caspase-1. Further experiments revealed that S1-induced increase in the production of TNFα, IL-6, IL-1β and IL-8 was reduced in the presence of BAY11-7082 and SKF 86002, while CRID3 pre-treatment resulted in the reduction of IL-1β production. These results suggest that SARS-CoV-2 spike glycoprotein S1 stimulated PBMCs to release pro-inflammatory cytokines through mechanisms involving activation of NF-κB, p38 MAPK and NLRP3 inflammasome. It is proposed that the clinical benefits of dexamethasone in COVID-19 are possibly due to its anti-inflammatory activity in reducing SARS-CoV-2 cytokine storm.
Collapse
|