51
|
Severity of COVID-19 patients with coexistence of asthma and vitamin D deficiency. INFORMATICS IN MEDICINE UNLOCKED 2022; 34:101116. [PMID: 36338941 PMCID: PMC9616486 DOI: 10.1016/j.imu.2022.101116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19)-driven global pandemic triggered innumerable health complications, imposing great challenges in managing other respiratory diseases like asthma. Furthermore, increases in the underlying inflammation involved in the fatality of COVID-19 have been linked with lack of vitamin D. In this research work, we intend to investigate the possible genetic linkage of asthma and vitamin D deficiency with the severity and fatality of COVID-19 using a network-based approach. We identified and analysed 41 and 14 differentially expressed genes (DEGs) of COVID-19 being common with asthma and vitamin D deficiency, respectively, through the comparative differential gene expression analysis and their footprints on signalling pathways. Gene set enrichment analysis for GO terms and signalling pathways reveals key biological activities, including inflammatory response-related pathways (e.g., cytokine- and chemokine-mediated signalling pathways, IL-17, and TNF signalling pathways). Besides, the Protein–Protein Interaction network analysis of those DEGs reveals hub proteins, some of which are reported as inflammatory antiviral interferon-stimulated biomarkers that potentially drive the cytokine storm leading to COVID-19 severity and fatality, and contributes in the early stage of viral replication, respectively. Moreover, the regulatory network analysis found these DEGs associated with antiviral and tumour inhibitory transcription factors and micro-RNAs. Finally, drug–target enrichment analysis yields tetradioxin, estradiol, arsenenous acid, and zinc, which have been reported to be effective in suppressing the pro-inflammatory cytokines production, and other respiratory tract infections. Our results yield shared biomarker-driven key hypotheses followed by network-based analytics, demystifying the mechanistic details of COVID-19 comorbidity of asthma and vitamin D deficiency with their potential therapeutic implications.
Collapse
|
52
|
Sanabria-Diaz G, Etter MM, Melie-Garcia L, Lieb JM, Psychogios MN, Hutter G, Granziera C. Brain cortical alterations in COVID-19 patients with neurological symptoms. Front Neurosci 2022; 16:992165. [PMID: 36340780 PMCID: PMC9630324 DOI: 10.3389/fnins.2022.992165] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/28/2022] [Indexed: 07/29/2023] Open
Abstract
Background Growing evidence suggests that the central nervous system is affected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), since infected patients suffer from acute and long-term neurological sequelae. Nevertheless, it is currently unknown whether the virus affects the brain cortex. The purpose of this study was to assess the cortical gray matter volume, the cortical thickness, and the cortical surface area in a group of SARS-CoV-2 infected patients with neurological symptoms compared to healthy control subjects. Additionally, we analyzed the cortical features and the association with inflammatory biomarkers in the cerebrospinal fluid (CSF) and plasma. Materials and methods Thirty-three patients were selected from a prospective cross-sectional study cohort during the ongoing pandemic (August 2020-April 2021) at the university hospitals of Basel and Zurich (Switzerland). The group included patients with different neurological symptom severity (Class I: nearly asymptomatic/mild symptoms, II: moderate symptoms, III: severe symptoms). Thirty-three healthy age and sex-matched subjects that underwent the same MRI protocol served as controls. For each anatomical T1w MPRAGE image, regional cortical gray matter volume, thickness, and surface area were computed with FreeSurfer. Using a linear regression model, cortical measures were compared between groups (patients vs. controls; Class I vs. II-III), with age, sex, MRI magnetic field strength, and total intracranial volume/mean thickness/total surface area as covariates. In a subgroup of patients, the association between cortical features and clinical parameters was assessed using partial correlation adjusting for the same covariates. P-values were corrected using a false discovery rate (FDR). Results Our findings revealed a lower cortical volume in COVID-19 patients' orbitofrontal, frontal, and cingulate regions than in controls (p < 0.05). Regional gray matter volume and thickness decreases were negatively associated with CSF total protein levels, the CSF/blood-albumin ratio, and CSF EN-RAGE levels. Conclusion Our data suggest that viral-triggered inflammation leads to neurotoxic damage in some cortical areas during the acute phase of a COVID-19 infection in patients with neurological symptoms.
Collapse
Affiliation(s)
- Gretel Sanabria-Diaz
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Manina Maja Etter
- Brain Tumor Immunotherapy Lab, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Division of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Lester Melie-Garcia
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Johanna M. Lieb
- Department of Neuroradiology, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, Basel, Switzerland
| | - Marios-Nikos Psychogios
- Department of Neuroradiology, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, Basel, Switzerland
| | - Gregor Hutter
- Brain Tumor Immunotherapy Lab, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Division of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Cristina Granziera
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
53
|
Tanveer A, Akhtar B, Sharif A, Saleem U, Rasul A, Ahmad A, Jilani K. Pathogenic role of cytokines in COVID-19, its association with contributing co-morbidities and possible therapeutic regimens. Inflammopharmacology 2022; 30:1503-1516. [PMID: 35948809 PMCID: PMC9365214 DOI: 10.1007/s10787-022-01040-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/15/2022]
Abstract
The Covid-19, a threatening pandemic, was originated from China in December 2019 and spread quickly to all over the world. The pathogenesis of coronavirus is linked with the disproportionate response of the immune system. This involves the systemic inflammatory reaction which is characterized by marked pro-inflammatory cytokine release commonly known as cytokine release storm (CRS). The pro inflammatory cytokines are involved in cascade of pulmonary inflammation, hyper coagulation and thrombosis which may be lethal for the individual. That's why, it is very important to have understanding of pro inflammatory cytokines and their pathological role in SARS-CoV-2. The pathogenesis of Covid is not the same in every individual, it can vary due to the presence of pre-existing comorbidities like suffering from already an inflammatory disease such as rheumatoid arthritis (RA), inflammatory bowel disease (IBD), chronic obstructive pulmonary disease (COPD), an immune-compromised patients suffering from Diabetes Mellitus (DM) and Tuberculosis (TB) are more vulnerable morbidity and complications following COVID-19. This review is particularly related to COVID-19 patients having comorbidity of other inflammatory diseases. We have discussed the brief pathogenesis of COVID-19 and cytokines release storm with reference to other co-morbidities including RA, IBD, COPD, DM and TB. The available therapeutic regimens for COVID-19 including cytokine inhibitors, anti-viral, anti-biotic, bronchodilators, JAK- inhibitors, immunomodulators and anti-fibrotic agents have also been discussed briefly. Moreover, newly emerging medicines in the clinical trials have also been discussed which are found to be effective in treating Covid-19.
Collapse
Affiliation(s)
- Ayesha Tanveer
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Bushra Akhtar
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan.
| | - Ali Sharif
- Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore, Pakistan
| | - Uzma Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Aftab Ahmad
- Department of Biochemistry, University of Agriculture, Faisalabad, Pakistan
- Center of Advanced Studies in Agriculture and Food Security (CAS-AFS), University of Agriculture, Faisalabad, Pakistan
| | - Kashif Jilani
- Department of Biochemistry, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
54
|
Lu LW, Gao Y, Quek SY, Foster M, Eason CT, Liu M, Wang M, Chen JH, Chen F. The landscape of potential health benefits of carotenoids as natural supportive therapeutics in protecting against Coronavirus infection. Biomed Pharmacother 2022; 154:113625. [PMID: 36058151 PMCID: PMC9428603 DOI: 10.1016/j.biopha.2022.113625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 01/08/2023] Open
Abstract
The Coronavirus Disease-2019 (COVID-19) pandemic urges researching possibilities for prevention and management of the effects of the virus. Carotenoids are natural phytochemicals of anti-oxidant, anti-inflammatory and immunomodulatory properties and may exert potential in aiding in combatting the pandemic. This review presents the direct and indirect evidence of the health benefits of carotenoids and derivatives based on in vitro and in vivo studies, human clinical trials and epidemiological studies and proposes possible mechanisms of action via which carotenoids may have the capacity to protect against COVID-19 effects. The current evidence provides a rationale for considering carotenoids as natural supportive nutrients via antioxidant activities, including scavenging lipid-soluble radicals, reducing hypoxia-associated superoxide by activating antioxidant enzymes, or suppressing enzymes that produce reactive oxygen species (ROS). Carotenoids may regulate COVID-19 induced over-production of pro-inflammatory cytokines, chemokines, pro-inflammatory enzymes and adhesion molecules by nuclear factor kappa B (NF-κB), renin-angiotensin-aldosterone system (RAS) and interleukins-6- Janus kinase-signal transducer and activator of transcription (IL-6-JAK/STAT) pathways and suppress the polarization of pro-inflammatory M1 macrophage. Moreover, carotenoids may modulate the peroxisome proliferator-activated receptors γ by acting as agonists to alleviate COVID-19 symptoms. They also may potentially block the cellular receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), human angiotensin-converting enzyme 2 (ACE2). These activities may reduce the severity of COVID-19 and flu-like diseases. Thus, carotenoid supplementation may aid in combatting the pandemic, as well as seasonal flu. However, further in vitro, in vivo and in particular long-term clinical trials in COVID-19 patients are needed to evaluate this hypothesis.
Collapse
|
55
|
Han W, Cheng Y, Chen J, Zhong H, Hu Z, Chen S, Zong L, Hong L, Chan TF, King I, Gao X, Li Y. Self-supervised contrastive learning for integrative single cell RNA-seq data analysis. Brief Bioinform 2022; 23:bbac377. [PMID: 36089561 PMCID: PMC9487595 DOI: 10.1093/bib/bbac377] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/20/2022] [Indexed: 12/14/2022] Open
Abstract
We present a novel self-supervised Contrastive LEArning framework for single-cell ribonucleic acid (RNA)-sequencing (CLEAR) data representation and the downstream analysis. Compared with current methods, CLEAR overcomes the heterogeneity of the experimental data with a specifically designed representation learning task and thus can handle batch effects and dropout events simultaneously. It achieves superior performance on a broad range of fundamental tasks, including clustering, visualization, dropout correction, batch effect removal, and pseudo-time inference. The proposed method successfully identifies and illustrates inflammatory-related mechanisms in a COVID-19 disease study with 43 695 single cells from peripheral blood mononuclear cells.
Collapse
Affiliation(s)
- Wenkai Han
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Yuqi Cheng
- Department of Computer Science and Engineering (CSE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jiayang Chen
- Department of Computer Science and Engineering (CSE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Huawen Zhong
- Biological and Environmental Sciences & Engineering Division (BESE), Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Zhihang Hu
- Department of Computer Science and Engineering (CSE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Siyuan Chen
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Licheng Zong
- Department of Computer Science and Engineering (CSE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Liang Hong
- Department of Computer Science and Engineering (CSE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Ting-Fung Chan
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Irwin King
- Department of Computer Science and Engineering (CSE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
- BioMap, Beijing, China
| | - Yu Li
- Department of Computer Science and Engineering (CSE), The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- The CUHK Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen, 518057, China
| |
Collapse
|
56
|
Lan Y, Wang H, Wu J, Meng X. Cytokine storm-calming property of the isoquinoline alkaloids in Coptis chinensis Franch. Front Pharmacol 2022; 13:973587. [PMID: 36147356 PMCID: PMC9485943 DOI: 10.3389/fphar.2022.973587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/16/2022] [Indexed: 11/18/2022] Open
Abstract
Coronavirus disease (COVID-19) has spread worldwide and its effects have been more devastating than any other infectious disease. Importantly, patients with severe COVID-19 show conspicuous increases in cytokines, including interleukin (IL)-6, monocyte chemoattractant protein (MCP)-1, IL-8, tumor necrosis factor (TNF)-α, IL-1, IL-18, and IL-17, with characteristics of the cytokine storm (CS). Although recently studied cytokine inhibitors are considered as potent and targeted approaches, once an immunological complication like CS happens, anti-viral or anti-inflammation based monotherapy alone is not enough. Interestingly, certain isoquinoline alkaloids in Coptis chinensis Franch. (CCFIAs) exerted a multitude of biological activities such as anti-inflammatory, antioxidant, antibacterial, and immunomodulatory etc, revealing a great potential for calming CS. Therefore, in this timeline review, we report and compare the effects of CCFIAs to attenuate the exacerbation of inflammatory responses by modulating signaling pathways like NF-ĸB, mitogen-activated protein kinase, JAK/STAT, and NLRP3. In addition, we also discuss the role of berberine (BBR) in two different triggers of CS, namely sepsis and viral infections, as well as its clinical applications. These evidence provide a rationale for considering CCFIAs as therapeutic agents against inflammatory CS and this suggestion requires further validation with clinical studies.
Collapse
Affiliation(s)
- Yuejia Lan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiasi Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Jiasi Wu, ; Xianli Meng,
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Jiasi Wu, ; Xianli Meng,
| |
Collapse
|
57
|
Zinellu A, Mangoni AA. A systematic review and meta-analysis of the association between the neutrophil, lymphocyte, and platelet count, neutrophil-to-lymphocyte ratio, and platelet-to-lymphocyte ratio and COVID-19 progression and mortality. Expert Rev Clin Immunol 2022; 18:1187-1202. [PMID: 36047369 DOI: 10.1080/1744666x.2022.2120472] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Severe manifestations of coronavirus disease 2019 (COVID-19) are associated with alterations in blood cells that regulate immunity, inflammation, and hemostasis. We conducted an updated systematic review and meta-analysis of the association between the neutrophil, lymphocyte, and platelet count, neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR), and COVID-19 progression and mortality. METHODS A systematic literature search was conducted in PubMed, Web of Science, and Scopus for studies published between January 2020 and June 2022. RESULTS In 71 studies reporting the investigated parameters within 48 hours of admission, higher NLR (HR 1.21, 95% CI 1.16 to 1.27, p < 0.0001), relative neutrophilia (HR 1.62, 95% CI 1.46 to 1.80, p < 0.0001), relative lymphopenia (HR 1.62, 95% CI 1.27 to 2.08, p < 0.001), and relative thrombocytopenia (HR 1.74, 95% CI 1.36 to 2.22, p < 0.001), but not PLR (p = 0.11), were significantly associated with disease progression and mortality. Between-study heterogeneity was large-to-extreme. The magnitude and direction of the effect size were not modified in sensitivity analysis. CONCLUSIONS NLR and neutrophil, lymphocyte, and platelet count significantly discriminate COVID-19 patients with different progression and survival outcomes. (PROSPERO registration number: CRD42021267875).
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.,Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
58
|
Chen S, Liu Y, Ge J, Yin J, Shi T, Ntambara J, Cheng Z, Chu M, Gu H. Tetrandrine Treatment May Improve Clinical Outcome in Patients with COVID-19. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58091194. [PMID: 36143871 PMCID: PMC9503147 DOI: 10.3390/medicina58091194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/23/2022] [Accepted: 08/27/2022] [Indexed: 12/02/2022]
Abstract
Background and objectives: The COVID-19 pandemic continues worldwide, and there is no effective treatment to treat it. Chinese medicine is considered the recommended treatment for COVID-19 in China. This study aimed to examine the effectiveness of tetrandrine in treating COVID-19, which is originally derived from Chinese medicine. Materials and Methods: A total of 60 patients, categorized into three types (mild, moderate, severe), from Daye Hospital of Chinese Medicine with a diagnosis of COVID-19 were included in this study. Demographics, medical history, treatment, and results were collected. We defined two main groups according to the clinical outcome between improvement and recovery. All underlying factors including clinical outcomes were assessed in the total number of COVID-19 patients and moderate-type patients. Results: In a total of 60 patients, there were significant differences in the clinical outcome underlying treatment with antibiotics, tetrandrine, and arbidol (p < 0.05). When the comparison was limited to the moderate type, treatment with tetrandrine further increased recovery rate (p = 0.007). However, the difference disappeared, and no association was indicated between the clinical outcome and the treatment with and without antibiotic (p = 0.224) and arbidol (p = 0.318) in the moderate-type patients. In all-type and moderate-type patients, tetrandrine improved the rate of improvement in cough and fatigue on day 7 (p < 0.05). Conclusions: Tetrandrine may improve clinical outcome in COVID-19 patientsand could be a promising potential natural antiviral agent for the prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Shiyin Chen
- School of Medicine, Nantong University, Nantong 226000, China
| | - Yiran Liu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong 226000, China
| | - Juan Ge
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), Nantong 226000, China
| | - Jianzhong Yin
- Department of Respiratory, Daye Hospital of Chinese Medicine, Daye 435100, China
| | - Ting Shi
- Department of Respiratory, Daye Hospital of Chinese Medicine, Daye 435100, China
| | - James Ntambara
- Department of Epidemiology, School of Public Health, Nantong University, Nantong 226000, China
| | - Zhounan Cheng
- Department of Epidemiology, School of Public Health, Nantong University, Nantong 226000, China
| | - Minjie Chu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong 226000, China
- Correspondence: (M.C.); (H.G.)
| | - Hongyan Gu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), Nantong 226000, China
- Correspondence: (M.C.); (H.G.)
| |
Collapse
|
59
|
Study on the potential of Sanghuangporus sanghuang and its components as COVID-19 spike protein receptor binding domain inhibitors. Biomed Pharmacother 2022; 153:113434. [PMID: 36076488 PMCID: PMC9288968 DOI: 10.1016/j.biopha.2022.113434] [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: 05/17/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 01/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has led to the most severe global pandemic, which began in Wuhan, China. Angiotensin-converting enzyme 2 (ACE2) combines with the spike protein of SARS-CoV-2, allowing the virus to cross the membrane and enter the cell. SARS-CoV-2 is modified by the transmembrane protease serine 2 (TMPRSS2) to facilitate access to cells. Accordingly, ACE2 and TMPRSS2 are targets of vital importance for the avoidance of SARS-CoV-2 infection. Sanghuangporus sanghuang (SS) is a traditional Chinese medicine that has been demonstrated to have antitumor, antioxidant, anti-inflammatory, antidiabetic, hepatoprotective, neuroprotective and immunomodulatory properties. In this paper, we demonstrated that SS decreased ACE2 and TMPRSS2 expression in cell lines and a mouse model without cytotoxicity or organ damage. Liver and kidney sections were confirmed to have reduced expression of ACE2 and TMPRSS2 by immunohistochemistry (IHC) assessment. Then, hispidin, DBA, PAC, PAD and CA, phenolic compounds of SS, were also tested and verified to reduce the expression of ACE2 and TMPRSS2. In summary, the results indicate that SS and its phenolic compounds have latent capacity for preventing SARS-CoV-2 infection in the future.
Collapse
|
60
|
Zavalaga-Zegarra HJ, Palomino-Gutierrez JJ, Ulloque-Badaracco JR, Mosquera-Rojas MD, Hernandez-Bustamante EA, Alarcon-Braga EA, Benites-Zapata VA, Herrera-Añazco P, Hernandez AV. C-Reactive Protein-to-Albumin Ratio and Clinical Outcomes in COVID-19 Patients: A Systematic Review and Meta-Analysis. Trop Med Infect Dis 2022; 7:186. [PMID: 36006278 PMCID: PMC9414550 DOI: 10.3390/tropicalmed7080186] [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: 07/08/2022] [Revised: 08/01/2022] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
C-reactive protein-to-albumin ratio (CAR) is an independent risk factor in cardiovascular, cerebrovascular, and infectious diseases. Through this study, we investigated the CAR values with respect to the severity and mortality of COVID-19 patients. We performed a systematic review and meta-analysis to retrieve studies that evaluated CAR values upon hospital admission in relation to the severity or mortality of COVID-19 patients. We adopted a random-effect model to calculate the pooled mean difference (MD) and their 95% confidence intervals (CI). Quality assessment was appraised using a Newcastle−Ottawa scale and publication bias was assessed using the Begg-test and funnel plot. We equally performed a subgroup analysis using study location and a sensitivity analysis only with studies with low risk of bias. We analyzed 32 studies (n = 12445). Severe COVID-19 patients had higher on-admission CAR values than non-severe COVID-19 patients (MD: 1.69; 95% CI: 1.35−2.03; p < 0.001; I2 = 89%). Non-survivor patients with COVID-19 had higher CAR values than survivor patients (MD: 2.59; 95% CI: 1.95−3.23; p < 0.001; I2 = 92%). In sensitivity analysis, the relationship remained with a decreasing of heterogeneity for severity (MD: 1.22; 95% CI: 1.03−1.40; p < 0.001; I2 = 13%) and for mortality (MD: 2.99; 95% CI: 2.47−3.51; p < 0.001; I2 = 0%). High CAR values were found in COVID-19 patients who developed severe disease or died.
Collapse
Affiliation(s)
| | | | - Juan R. Ulloque-Badaracco
- Escuela de Medicina, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
- Sociedad Científica de Estudiantes de Medicina de la Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Melany D. Mosquera-Rojas
- Escuela de Medicina, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
- Sociedad Científica de Estudiantes de Medicina de la Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Enrique A. Hernandez-Bustamante
- Sociedad Científica de Estudiantes de Medicina de la Universidad Nacional de Trujillo, Trujillo 13011, Peru
- Grupo Peruano de Investigación Epidemiológica, Unidad para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, Lima 15012, Peru
| | - Esteban A. Alarcon-Braga
- Escuela de Medicina, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
- Sociedad Científica de Estudiantes de Medicina de la Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Vicente A. Benites-Zapata
- Unidad de Investigación para la Generación y Síntesis de Evidencias en Salud, Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima 15012, Peru
| | - Percy Herrera-Añazco
- Escuela de Enfermería, Universidad Privada San Juan Bautista, Lima 15067, Peru
- Instituto de Evaluación de Tecnologías en Salud e Investigación—IETSI, EsSalud, Lima 14072, Peru
| | - Adrian V. Hernandez
- Unidad de Revisiones Sistemáticas y Meta-Análisis, Guías de Práctica Clínica y Evaluaciones de Tecnología Sanitaria, Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima 15012, Peru
- Health Outcomes, Policy, and Evidence Synthesis (HOPES) Group, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
61
|
Hosseinzadeh A, Bagherifard A, Koosha F, Amiri S, Karimi-Behnagh A, Reiter RJ, Mehrzadi S. Melatonin effect on platelets and coagulation: Implications for a prophylactic indication in COVID-19. Life Sci 2022; 307:120866. [PMID: 35944663 PMCID: PMC9356576 DOI: 10.1016/j.lfs.2022.120866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 11/04/2022]
Abstract
Severe COVID-19 is associated with the dynamic changes in coagulation parameters. Coagulopathy is considered as a major extra-pulmonary risk factor for severity and mortality of COVID-19; patients with elevated levels of coagulation biomarkers have poorer in-hospital outcomes. Oxidative stress, alterations in the activity of cytochrome P450 enzymes, development of the cytokine storm and inflammation, endothelial dysfunction, angiotensin-converting enzyme 2 (ACE2) enzyme malfunction and renin–angiotensin system (RAS) imbalance are among other mechanisms suggested to be involved in the coagulopathy induced by severe acute respiratory syndrome coronavirus (SARS-CoV-2). The activity and function of coagulation factors are reported to have a circadian component. Melatonin, a multipotential neurohormone secreted by the pineal gland exclusively at night, regulates the cytokine system and the coagulation cascade in infections such as those caused by coronaviruses. Herein, we review the mechanisms and beneficial effects of melatonin against coagulopathy induced by SARS-CoV-2 infection.
Collapse
|
62
|
Attisani L, Bissacco D, Pucci A, Luoni G, Luzzani L, Pegorer MA, Settembrini AM, Wohlauer MV, Bellosta R. COVID-19 and limb ischemia: experience first. THE JOURNAL OF CARDIOVASCULAR SURGERY 2022; 63:537-538. [PMID: 35848872 DOI: 10.23736/s0021-9509.22.12314-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Affiliation(s)
- Luca Attisani
- Vascular and Endovascular Surgery Unit, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Daniele Bissacco
- Vascular Surgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy -
| | - Alessandro Pucci
- Vascular and Endovascular Surgery Unit, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Giorgio Luoni
- Vascular and Endovascular Surgery Unit, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Luca Luzzani
- Vascular and Endovascular Surgery Unit, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Matteo A Pegorer
- Vascular and Endovascular Surgery Unit, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Alberto M Settembrini
- Vascular Surgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Max V Wohlauer
- Department of Vascular Surgery, University of Colorado, Denver, CO, USA
| | - Raffaello Bellosta
- Vascular and Endovascular Surgery Unit, Poliambulanza Foundation Hospital, Brescia, Italy
| |
Collapse
|
63
|
Fibrinogen-to-Albumin Ratio and Blood Urea Nitrogen-to-Albumin Ratio in COVID-19 Patients: A Systematic Review and Meta-Analysis. Trop Med Infect Dis 2022; 7:tropicalmed7080150. [PMID: 36006242 PMCID: PMC9414552 DOI: 10.3390/tropicalmed7080150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/08/2023] Open
Abstract
Fibrinogen-to-albumin ratio (FAR) and blood urea nitrogen-to-albumin ratio (BAR) are inflammatory biomarkers that have been associated with clinical outcomes of multiple diseases. The objective of this study is to evaluate the association of these biomarkers with the severity and mortality of COVID-19 patients. A systematic search was performed in five databases. Observational studies that reported the association between FAR and BAR values with the severity and mortality of COVID-19 patients were included. Random-effects models were used for meta-analyses, and effects were expressed as Odds Ratio (OR) and their 95% confidence intervals (CI). Publication bias was assessed using the Begg test, while the quality assessment was assessed using the Newcastle Ottawa Scale. A total of 21 studies (n = 7949) were included. High FAR values were associated with a higher risk of severity (OR: 2.41; 95% CI 1.41−4.12; p < 0.001) and mortality (OR: 2.05; 95% CI 1.66−2.54; p < 0.001). High BAR values were associated with higher risk of mortality (OR: 4.63; 95% CI 2.11−10.15; p < 0.001). However, no statistically significant association was found between BAR values and the risk of severity (OR: 1.16; 95% CI 0.83−1.63; p = 0.38). High FAR and BAR values were associated with poor clinical outcomes.
Collapse
|
64
|
Feng Q, Si Y, Zhu L, Wang F, Fang J, Pan C, Gao X, Liu W. Anti-inflammatory effects of a SERP 30 polysaccharide from the residue of Sarcandra glabra against lipopolysaccharide-induced acute respiratory distress syndrome in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115262. [PMID: 35398243 DOI: 10.1016/j.jep.2022.115262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sarcandra glabra (Thunb.) Nakai, a valuable dietetic Chinese herb, is still widely used today. Multiple ingredients of S. glabra with a variety of activities such as anti-inflammatory, antiviral, and antitumor were studied. However, the Sarcandra glabra (Thunb.) Nakai polysaccharide hasn't been reported for its anti-inflammatory effect. AIM OF THE STUDY In this study, the anti-inflammatory activity of Sarcandra glabra (Thunb.) Nakai polysaccharide was assessed in LPS-induced ARDS mice. MATERIALS AND METHODS A polysaccharide coded as SERP 30 was obtained by water extraction, alcohol precipitation, and gel filtration. After the physicochemical properties determination and structural characterization, LPS induced-mice ARDS model was used to evaluate the anti-inflammatory and associated antioxidant activities of SERP 30. H&E staining was used to observe the seriousness of lung injury in mice. The ELISA method was used to measure the expression of inflammatory factors (TNF-α and IL-6) in the serum of the mice. The TBA method and the WST-1 method were used to evaluate the oxidative stress injury. Immunohistochemistry was used to distinguish the expression of metalloproteinase-9 (MMP-9), heparinase (HPA), syndecan-1, and decorin in ARDS-mice lung tissue. Western blotting was used to confirm the expression of related proteins in mouse lung tissue. RESULTS SERP 30 had a potential role in improving lung damage, reducing inflammation, and preventing oxidative stress. Moreover, SERP 30 significantly attenuated the damage to the endothelial glycocalyx and maintained the integrity of the glycocalyx. The western blotting result implied that the main anti-inflammatory mechanism is directed towards NF-κB and MAPK signaling pathways with inhibiting the activation of associated proteins. CONCLUSION This research provides a theoretical basis for treating ARDS by using a byproduct from food resource.
Collapse
Affiliation(s)
- Qi Feng
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yu Si
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Lingling Zhu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Feng Wang
- Simcere Pharmaceutical Group Limited, Nanjing, 210042, PR China
| | - Junqiang Fang
- Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Jinan, 250000, PR China
| | - Chun Pan
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, PR China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Wei Liu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
65
|
Veronesi F, Contartese D, Martini L, Visani A, Fini M. Speculation on the pathophysiology of musculoskeletal injury with COVID-19 infection. Front Med (Lausanne) 2022; 9:930789. [PMID: 35911401 PMCID: PMC9329661 DOI: 10.3389/fmed.2022.930789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/28/2022] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) primarily affects the respiratory tract, but also many other organs and tissues, leading to different pathological pictures, such as those of the musculoskeletal tissues. The present study should be considered as a speculation on the relationship between COVID-19 infection and some frequent musculoskeletal pathologies, in particular sarcopenia, bone loss/osteoporosis (OP) and fracture risk and osteoarthritis (OA), to hypothesize how the virus acts on these pathologies and consequently on the tissue regeneration/healing potential. The study focuses in particular on the modalities of interaction of COVID-19 with Angiotensin-Converting Enzyme 2 (ACE2) and on the “cytokine storm.” Knowing the effects of COVID-19 on musculoskeletal tissues could be important also to understand if tissue regenerative/reparative capacity is compromised, especially in elderly and frail patients. We speculate that ACE2 and serine proteases together with an intense inflammation, immobilization and malnutrition could be the responsible for muscle weakness, altered bone remodeling, increase in bone fracture risk and inflammatory joint pathologies. Future preclinical and clinical studies may focus on the regenerative/reparative properties of the musculoskeletal tissues after COVID-19 infection, toward a personalized treatment usually based on scaffolds, cells, and growth factors.
Collapse
|
66
|
Doan LH, Chu LW, Huang ZY, Nguyen AT, Lee CY, Huang CL, Chang YF, Hsieh WY, Nguyen TTH, Lin CH, Su CL, Chuang TH, Lai JM, Wang FS, Yang CJ, Liu HK, Ping YH, Huang CYF. Virofree, an Herbal Medicine-Based Formula, Interrupts the Viral Infection of Delta and Omicron Variants of SARS-CoV-2. Front Pharmacol 2022; 13:905197. [PMID: 35860023 PMCID: PMC9289459 DOI: 10.3389/fphar.2022.905197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/20/2022] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) remains a threat with the emergence of new variants, especially Delta and Omicron, without specific effective therapeutic drugs. The infection causes dysregulation of the immune system with a cytokine storm that eventually leads to fatal acute respiratory distress syndrome (ARDS) and further irreversible pulmonary fibrosis. Therefore, the promising way to inhibit infection is to disrupt the binding and fusion between the viral spike and the host ACE2 receptor. A transcriptome-based drug screening platform has been developed for COVID-19 to explore the possibility and potential of the long-established drugs or herbal medicines to reverse the unique genetic signature of COVID-19. In silico analysis showed that Virofree, an herbal medicine, reversed the genetic signature of COVID-19 and ARDS. Biochemical validations showed that Virofree could disrupt the binding of wild-type and Delta-variant spike proteins to ACE2 and its syncytial formation via cell-based pseudo-typed viral assays, as well as suppress binding between several variant recombinant spikes to ACE2, especially Delta and Omicron. Additionally, Virofree elevated miR-148b-5p levels, inhibited the main protease of SARS-CoV-2 (Mpro), and reduced LPS-induced TNF-α release. Virofree also prevented cellular iron accumulation leading to ferroptosis which occurs in SARS-CoV-2 patients. Furthermore, Virofree was able to reduce pulmonary fibrosis-related protein expression levels in vitro. In conclusion, Virofree was repurposed as a potential herbal medicine to combat COVID-19. This study highlights the inhibitory effect of Virofree on the entry of Delta and Omicron variants of SARS-CoV-2, which have not had any effective treatments during the emergence of the new variants spreading.
Collapse
Affiliation(s)
- Ly Hien Doan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Li-Wei Chu
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Zi-Yi Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- ASUS Intelligent Cloud Services, Taipei, Taiwan
| | - Anh Thuc Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Taiwan National Graduate Program in Molecular Medicine, Academia Sinica, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Yin Lee
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Chien-Ling Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | - Wen-Yu Hsieh
- Division of Basic Chinese Medicine, National Research Institute of Chinese Medicine (NRICM), Ministry of Health and Welfare, Taipei, Taiwan
| | - Trang Thi Huyen Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chao-Hsiung Lin
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Life Sciences and Institute of Genome Sciences, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Aging and Health Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Li Su
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Jin-Mei Lai
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Feng-Sheng Wang
- Department of Chemical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - Chia-Jui Yang
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Hui-Kang Liu
- Division of Basic Chinese Medicine, National Research Institute of Chinese Medicine (NRICM), Ministry of Health and Welfare, Taipei, Taiwan
- Ph.D. Program in the Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yueh-Hsin Ping
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biophotonics, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Taiwan National Graduate Program in Molecular Medicine, Academia Sinica, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
67
|
Rasmi Y, Hatamkhani S, Naderi R, Shokati A, Nayeb Zadeh V, Hosseinzadeh F, Farnamian Y, Jalali L. Molecular signaling pathways, pathophysiological features in various organs, and treatment strategies in SARS-CoV2 infection. Acta Histochem 2022; 124:151908. [PMID: 35662001 PMCID: PMC9130726 DOI: 10.1016/j.acthis.2022.151908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/19/2022] [Indexed: 01/08/2023]
Abstract
Cytokine storms and extra-activated cytokine signaling pathways can lead to severe tissue damage and patient death. Activation of inflammatory signaling pathways during Cytokine storms are an important factor in the development of acute respiratory syndrome (SARS-CoV-2), which is the major health problem today, causing systemic and local inflammation. Cytokine storms attract many inflammatory cells that attack the lungs and other organs and cause tissue damage. Angiotensin-converting enzyme 2 (ACE2) are expressed in a different type of tissues. inhibition of ACE2 activity impairs renin-angiotensin (RAS) function, which is related to the severity of symptoms and mortality rate in COVID-19 patients. Different signaling cascades are activated, affecting various organs during SARS-CoV-2 infection. Nowadays, there is no specific treatment for COVID-19, but scientists have recognized and proposed several treatment alternatives, including applying cytokine inhibitors, immunomodulators, and plasma therapy. Herein, we have provided the detailed mechanism of SARS-CoV-2 induced cytokine signaling and its connection with pathophysiological features in different organs. Possible treatment options to cope with the severe clinical manifestations of COVID-19 are also discussed.
Collapse
Affiliation(s)
- Yousef Rasmi
- Cellular and Molecular Research Center,Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shima Hatamkhani
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran; Department of Clinical Pharmacy, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Roya Naderi
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Physiology, school of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ameneh Shokati
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | | | - Faezeh Hosseinzadeh
- Department of Tissue Engineering, Qom University of Medical Sciences, Qom, Iran
| | - Yeganeh Farnamian
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Ladan Jalali
- Cellular and Molecular Research Center,Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
68
|
Agrawal P, Sambaturu N, Olgun G, Hannenhalli S. A Path-Based Analysis of Infected Cell Line and COVID-19 Patient Transcriptome Reveals Novel Potential Targets and Drugs Against SARS-CoV-2. Front Immunol 2022; 13:918817. [PMID: 35844595 PMCID: PMC9284228 DOI: 10.3389/fimmu.2022.918817] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Most transcriptomic studies of SARS-CoV-2 infection have focused on differentially expressed genes, which do not necessarily reveal the genes mediating the transcriptomic changes. In contrast, exploiting curated biological network, our PathExt tool identifies central genes from the differentially active paths mediating global transcriptomic response. Here we apply PathExt to multiple cell line infection models of SARS-CoV-2 and other viruses, as well as to COVID-19 patient-derived PBMCs. The central genes mediating SARS-CoV-2 response in cell lines were uniquely enriched for ATP metabolic process, G1/S transition, leukocyte activation and migration. In contrast, PBMC response reveals dysregulated cell-cycle processes. In PBMC, the most frequently central genes are associated with COVID-19 severity. Importantly, relative to differential genes, PathExt-identified genes show greater concordance with several benchmark anti-COVID-19 target gene sets. We propose six novel anti-SARS-CoV-2 targets ADCY2, ADSL, OCRL, TIAM1, PBK, and BUB1, and potential drugs targeting these genes, such as Bemcentinib, Phthalocyanine, and Conivaptan.
Collapse
Affiliation(s)
- Piyush Agrawal
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Narmada Sambaturu
- IISc Mathematics Initiative, Indian Institute of Science, Bangalore, India
| | - Gulden Olgun
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
69
|
Behl T, Kumar S, Singh S, Bhatia S, Albarrati A, Albratty M, Meraya AM, Najmi A, Bungau S. Reviving the mutual impact of SARS-COV-2 and obesity on patients: From morbidity to mortality. Biomed Pharmacother 2022; 151:113178. [PMID: 35644117 PMCID: PMC9127128 DOI: 10.1016/j.biopha.2022.113178] [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: 04/28/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 01/25/2023] Open
Abstract
Obesity-related metabolic dysfunction, endothelium imbalance, chronic inflammation, immune dysregulation, and its comorbidities may all have a role in systemic inflammation, leading to the pulmonary fibrosis and cytokine storm, which leads to failure of lung function, which is a hallmark of severe SARS-CoV-2 infection. Obesity may also disrupt the function of mucociliary escalators and cooperation of epithelial cell's motile cilia in the airway, limiting the clearance of the coronavirus that causes severe acute respiratory syndrome (SARS-CoV-2). Adipose tissues in obese patients have a greater number of proteases and receptors for SARS-CoV-2 admittance, proposing that they could serve as an accelerator and reservoir for this virus, boosting immunological response and systemic inflammation. Lastly, anti-inflammatory cytokines such as anti-IL-6 and the infusion of mesenchymal stem cells could be used as a modulation therapy of immunity to help COVID-19 patients. Obesity, on the other hand, is linked to the progress of COVID-19 through a variety of molecular pathways, and obese people are part of the SARS-CoV-2 susceptible individuals, necessitating more protective measures.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Sachin Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman; School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ali Albarrati
- Rehabilitation Health Sciences College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Abdulkarim M Meraya
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania; Doctoral School of Biomedical Sciences, University of Oradea, Oradea, Romania.
| |
Collapse
|
70
|
How to Restore Oxidative Balance That Was Disrupted by SARS-CoV-2 Infection. Int J Mol Sci 2022; 23:ijms23126377. [PMID: 35742820 PMCID: PMC9223498 DOI: 10.3390/ijms23126377] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/04/2022] [Accepted: 06/05/2022] [Indexed: 12/17/2022] Open
Abstract
Coronavirus 2019 disease (COVID-19) is caused by different variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which emerged in December of 2019. COVID-19 pathogenesis is complex and involves a dysregulated renin angiotensin system. Severe courses of the disease are associated with a dysregulated immunological response known as cytokine storm. Many scientists have demonstrated that SARS-CoV-2 impacts oxidative homeostasis and stimulates the production of reactive oxygen species (ROS). In addition, the virus inhibits glutathione (GSH) and nuclear factor erythroid 2-related factor 2 (NRF2)-a major antioxidant which induces expression of protective proteins and prevents ROS damage. Furthermore, the virus stimulates NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasomes which play a significant role in inducing a cytokine storm. A variety of agents with antioxidant properties have shown beneficial effects in experimental and clinical studies of COVID-19. This review aims to present mechanisms of oxidative stress induced by SARS-CoV-2 and to discuss whether antioxidative drugs can counteract detrimental outcomes of a cytokine storm.
Collapse
|
71
|
Meilik R, Ben-Assayag H, Meilik A, Berliner S, Zeltser D, Shapira I, Rogowski O, Goldiner I, Shenhar-Tsarfaty S, Wasserman A. Sepsis Related Mortality Associated with an Inflammatory Burst in Patients Admitting to the Department of Internal Medicine with Apparently Normal C-Reactive Protein Concentration. J Clin Med 2022; 11:jcm11113151. [PMID: 35683538 PMCID: PMC9181046 DOI: 10.3390/jcm11113151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
Background: Patients who are admitted to the Department of Internal Medicine with apparently normal C-reactive protein (CRP) concentration impose a special challenge due the assumption that they might not harbor a severe and potentially lethal medical condition. Methods: A retrospective cohort of all patients who were admitted to the Department of Internal Medicine with a CRP concentration of ≤31.9 mg/L and had a second CRP test obtained within the next 24 h. Seven day mortality data were analyzed. Results: Overall, 3504 patients were analyzed with a mean first and second CRP of 8.8 (8.5) and 14.6 (21.6) mg/L, respectively. The seven day mortality increased from 1.8% in the first quartile of the first CRP to 7.5% in the fourth quartile of the first CRP (p < 0.0001) and from 0.6% in the first quartile of the second CRP to 9.5% in the fourth quartile of the second CRP test (p < 0.0001), suggesting a clear relation between the admission CRP and in hospital seven day mortality. Conclusions: An association exists between the quartiles of CRP and 7-day mortality as well as sepsis related cause of death. Furthermore, the CRP values 24 h after hospital admission improved the discrimination.
Collapse
Affiliation(s)
- Ronnie Meilik
- Department of Internal Medicine “C”, “D”, & “E”, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel; (R.M.); (H.B.-A.); (S.B.); (D.Z.); (I.S.); (O.R.); (A.W.)
| | - Hadas Ben-Assayag
- Department of Internal Medicine “C”, “D”, & “E”, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel; (R.M.); (H.B.-A.); (S.B.); (D.Z.); (I.S.); (O.R.); (A.W.)
| | - Ahuva Meilik
- Clinical Performances Research and Operational Unit, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel;
| | - Shlomo Berliner
- Department of Internal Medicine “C”, “D”, & “E”, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel; (R.M.); (H.B.-A.); (S.B.); (D.Z.); (I.S.); (O.R.); (A.W.)
| | - David Zeltser
- Department of Internal Medicine “C”, “D”, & “E”, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel; (R.M.); (H.B.-A.); (S.B.); (D.Z.); (I.S.); (O.R.); (A.W.)
| | - Itzhak Shapira
- Department of Internal Medicine “C”, “D”, & “E”, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel; (R.M.); (H.B.-A.); (S.B.); (D.Z.); (I.S.); (O.R.); (A.W.)
| | - Ori Rogowski
- Department of Internal Medicine “C”, “D”, & “E”, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel; (R.M.); (H.B.-A.); (S.B.); (D.Z.); (I.S.); (O.R.); (A.W.)
| | - Ilana Goldiner
- Laboratory Medicine, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel;
| | - Shani Shenhar-Tsarfaty
- Department of Internal Medicine “C”, “D”, & “E”, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel; (R.M.); (H.B.-A.); (S.B.); (D.Z.); (I.S.); (O.R.); (A.W.)
- Correspondence:
| | - Asaf Wasserman
- Department of Internal Medicine “C”, “D”, & “E”, Tel Aviv Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel; (R.M.); (H.B.-A.); (S.B.); (D.Z.); (I.S.); (O.R.); (A.W.)
| |
Collapse
|
72
|
Bahoosh SR, Shokoohinia Y, Eftekhari M. Glucosinolates and their hydrolysis products as potential nutraceuticals to combat cytokine storm in SARS-COV-2. DARU : JOURNAL OF FACULTY OF PHARMACY, TEHRAN UNIVERSITY OF MEDICAL SCIENCES 2022; 30:245-252. [PMID: 35112323 PMCID: PMC8809497 DOI: 10.1007/s40199-022-00435-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/23/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The high mortality rate in severe cases of COVID-19 is mainly due to the strong upregulation of cytokines, called a cytokine storm. Hyperinflammation and multiple organ failure comprise the main clinical features of a cytokine storm. Nrf2 is a transcription factor which regulates the expression of genes involved in immune and inflammatory processes. Furthermore, Nrf2, as a master regulator, controls the activity of NF-κB which binds to the promoter of many pro-inflammatory genes inducible of various inflammatory factors. Inhibition of Nrf2 response was recently demonstrated in biopsies from patients with COVID-19, and Nrf2 agonists inhibited SARS-CoV-2 replication across cell lines in vitro. Glucosinolates and their hydrolysis products have excellent anti-inflammatory and antioxidant effects via the Nrf2 activation pathway, reduction in the NF-κB activation, and subsequent reduced cytokines levels. CONCLUSION Accordingly, these compounds can be helpful in combating the cytokine storm associated with COVID-19.
Collapse
Affiliation(s)
- Saba Rahimi Bahoosh
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yalda Shokoohinia
- Ric Scalzo Institute for Botanical Research, Southwest College of Naturopathic Medicine, Tempe, AZ, 85282, USA
| | - Mahdieh Eftekhari
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, 6715847141, Kermanshah, Iran. .,Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, P.O.BOX.6714415153, Kermanshah, Iran.
| |
Collapse
|
73
|
Mufarrih SH, Qureshi NQ, Yunus R, Ngo D, Katz D, Krakower D, Bhambhani V, Quadir J, Solleveld P, Banner-Goodspeed V, Mahmood F, Matyal R. Influence of Increasing Age and Body Mass Index of Gender in COVID-19 Patients. J Womens Health (Larchmt) 2022; 31:779-786. [PMID: 35708572 PMCID: PMC10163441 DOI: 10.1089/jwh.2021.0615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: The impact of gender on outcomes in patients suffering from coronavirus disease 2019 (COVID-19) is frequently debated. However, the synchronous influence of additional risk factors is seldom mentioned. With increasing emphasis on identifying patients who are at risk of complications from COVID-19, we decided to conduct a retrospective review to assess the influence of age and body mass index (BMI) on gender-based differences in outcomes. Materials and Methods: A retrospective review of 1288 patients was conducted at a tertiary care hospital. Binary logistic regression was used to assess differences in risk factors and outcomes between genders. The associations between predictors and outcomes were described using odds ratios in tables, forest plots, and regression curves plotted using Sigma Plot. Results: Majority of patients were women (53.6% vs. 46.4%). Median BMI in men was higher than women (p = 0.003). Key predictors for all-cause morbidity/mortality in men were diabetes, chronic kidney disease, and regular use of angiotensin-converting enzyme inhibitors. In women, age >65 and regular use of inhaled steroid were additional risk factors. Men had a higher risk of acute respiratory distress syndrome (2.83 [1.70-4.70]), acute renal failure (1.96 [1.20-3.20]), and had a longer length of stay (0.11 [1.52]). Obesity has a stronger bearing on outcomes in women, and age has a more pronounced effect on outcomes in men. Conclusion: Extremes of BMI and older age are associated with worse outcomes in both men and women. Obesity has a stronger bearing on outcomes of COVID-19 infection in women, while the effect of older age on outcomes is more pronounced in men.
Collapse
Affiliation(s)
- Syed Hamza Mufarrih
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nada Qaisar Qureshi
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Rayaan Yunus
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Debby Ngo
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Katz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Douglas Krakower
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Victoria Bhambhani
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Juweria Quadir
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Patricia Solleveld
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Valerie Banner-Goodspeed
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Feroze Mahmood
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Robina Matyal
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
74
|
Gudowska-Sawczuk M, Mroczko B. The Role of Nuclear Factor Kappa B (NF-κB) in Development and Treatment of COVID-19: Review. Int J Mol Sci 2022; 23:ijms23095283. [PMID: 35563673 PMCID: PMC9101079 DOI: 10.3390/ijms23095283] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 01/27/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes Coronavirus Disease 19 (COVID-19), a disease that has affected more than 500 million people worldwide since the end of 2019. Due to its high complications and death rates, there is still a need to find the best therapy for SARS-CoV-2 infection. The dysregulation of the inflammatory response in COVID-19 plays a very important role in disease progression. It has been observed that abnormal activity of Nuclear Factor kappa B (NF-κB) is directly associated with, inter alia, increased synthesis of proinflammatory factors. Therefore, this review paper focuses on the functions of NF-κB in the development of SARS-CoV-2 infection and potential application of NF-κB inhibitors in COVID-19 immunotherapy. A comprehensive literature search was performed using the MEDLINE/PubMed database. In the current review, it is highlighted that NF-κB plays important functions in the modulation of an adaptive inflammatory response, including inducing the expression of proinflammatory genes. Increased activation of NF-κB in SARS-CoV-2 infection was observed. The association between NF-κB activation and the expression of SARS-CoV-2 structural and non-structural proteins were also reported. It was observed that modulation of NF-κB using, e.g., traditional Chinese medicine or glucocorticosteroids resulted in decreased synthesis of proinflammatory factors caused by SARS-CoV-2 infection. This review summarizes the role of NF-κB in COVID-19 and describes its potential immunotherapeutic target in treatment of SARS-CoV-2 infection. However, indisputably more studies involving patients with a severe course of COVID-19 are sorely needed.
Collapse
Affiliation(s)
- Monika Gudowska-Sawczuk
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland;
- Correspondence: ; Tel.: +48-85-831-8703
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
75
|
Jiménez D, Torres Arias M. Immunouniverse of SARS-CoV-2. Immunol Med 2022; 45:186-224. [PMID: 35502127 DOI: 10.1080/25785826.2022.2066251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
SARS-CoV-2 virus has become a global health problem that has caused millions of deaths worldwide. The infection can present with multiple clinical features ranging from asymptomatic or mildly symptomatic patients to patients with severe or critical illness that can even lead to death. Although the immune system plays an important role in pathogen control, SARS-CoV-2 can drive dysregulation of this response and trigger severe immunopathology. Exploring the mechanisms of the immune response involved in host defense against SARS-CoV-2 allows us to understand its immunopathogenesis and possibly detect features that can be used as potential therapies to eliminate the virus. The main objective of this review on SARS-CoV-2 is to highlight the interaction between the virus and the immune response. We explore the function and action of the immune system, the expression of molecules at the site of infection that cause hyperinflammation and hypercoagulation disorders, the factors leading to the development of pneumonia and subsequent severe acute respiratory distress syndrome which is the leading cause of death in patients with COVID-19.
Collapse
Affiliation(s)
- Dennis Jiménez
- Departamento de Ciencias de la Vida y Agricultura, Carrera de Ingeniería en Biotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Pichincha, Ecuador
| | - Marbel Torres Arias
- Departamento de Ciencias de la Vida y Agricultura, Carrera de Ingeniería en Biotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Pichincha, Ecuador.,Laboratorio de Inmunología y Virología, CENCINAT, GISAH, Universidad de las Fuerzas Armadas, Sangolquí, Pichincha, Ecuador
| |
Collapse
|
76
|
Niedźwiedzka-Rystwej P, Majchrzak A, Kurkowska S, Małkowska P, Sierawska O, Hrynkiewicz R, Parczewski M. Immune Signature of COVID-19: In-Depth Reasons and Consequences of the Cytokine Storm. Int J Mol Sci 2022; 23:4545. [PMID: 35562935 PMCID: PMC9105989 DOI: 10.3390/ijms23094545] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
In the beginning of the third year of the fight against COVID-19, the virus remains at least still one step ahead in the pandemic "war". The key reasons are evolving lineages and mutations, resulting in an increase of transmissibility and ability to evade immune system. However, from the immunologic point of view, the cytokine storm (CS) remains a poorly understood and difficult to combat culprit of the extended number of in-hospital admissions and deaths. It is not fully clear whether the cytokine release is a harmful result of suppression of the immune system or a positive reaction necessary to clear the virus. To develop methods of appropriate treatment and therefore decrease the mortality of the so-called COVID-19-CS, we need to look deeply inside its pathogenesis, which is the purpose of this review.
Collapse
Affiliation(s)
| | - Adam Majchrzak
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 71-455 Szczecin, Poland; (A.M.); (M.P.)
| | - Sara Kurkowska
- Department of Nuclear Medicine, Pomeranian Medical University, 71-252 Szczecin, Poland;
| | - Paulina Małkowska
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
- Doctoral School, University of Szczecin, 71-412 Szczecin, Poland
| | - Olga Sierawska
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
- Doctoral School, University of Szczecin, 71-412 Szczecin, Poland
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland; (P.M.); (O.S.); (R.H.)
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 71-455 Szczecin, Poland; (A.M.); (M.P.)
| |
Collapse
|
77
|
Yang X, Rutkovsky AC, Zhou J, Zhong Y, Reese J, Schnell T, Albrecht H, Owens WB, Nagarkatti PS, Nagarkatti M. Characterization of Altered Gene Expression and Histone Methylation in Peripheral Blood Mononuclear Cells Regulating Inflammation in COVID-19 Patients. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1968-1977. [PMID: 35379747 PMCID: PMC9012677 DOI: 10.4049/jimmunol.2101099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
Abstract
The pandemic of COVID-19 has caused >5 million deaths in the world. One of the leading causes of the severe form of COVID-19 is the production of massive amounts of proinflammatory cytokines. Epigenetic mechanisms, such as histone/DNA methylation, miRNA, and long noncoding RNA, are known to play important roles in the regulation of inflammation. In this study, we investigated if hospitalized COVID-19 patients exhibit alterations in epigenetic pathways in their PBMCs. We also compared gene expression profiles between healthy controls and COVID-19 patients. Despite individual variations, the expressions of many inflammation-related genes, such as arginase 1 and IL-1 receptor 2, were significantly upregulated in COVID-19 patients. We also found the expressions of coagulation-related genes Von Willebrand factor and protein S were altered in COVID-19 patients. The expression patterns of some genes, such as IL-1 receptor 2, correlated with their histone methylation marks. Pathway analysis indicated that most of those dysregulated genes were in the TGF-β, IL-1b, IL-6, and IL-17 pathways. A targeting pathway revealed that the majority of those altered genes were targets of dexamethasone, which is an approved drug for COVID-19 treatment. We also found that the expression of bone marrow kinase on chromosome X, a member of TEC family kinases, was increased in the PBMCs of COVID-19 patients. Interestingly, some inhibitors of TEC family kinases have been used to treat COVID-19. Overall, this study provides important information toward identifying potential biomarkers and therapeutic targets for COVID-19 disease.
Collapse
Affiliation(s)
- Xiaoming Yang
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC; and
| | - Alex C Rutkovsky
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC; and
| | - Juhua Zhou
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC; and
| | - Yin Zhong
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC; and
| | - Julian Reese
- Prisma Health Richland Hospital, School of Medicine, University of South Carolina, Columbia, SC
| | - Timothy Schnell
- Prisma Health Richland Hospital, School of Medicine, University of South Carolina, Columbia, SC
| | - Helmut Albrecht
- Prisma Health Richland Hospital, School of Medicine, University of South Carolina, Columbia, SC
| | - William B Owens
- Prisma Health Richland Hospital, School of Medicine, University of South Carolina, Columbia, SC
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC; and
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC; and
| |
Collapse
|
78
|
Yeh YC, Doan LH, Huang ZY, Chu LW, Shi TH, Lee YR, Wu CT, Lin CH, Chiang ST, Liu HK, Chuang TH, Ping YH, Liu HS, Huang CYF. Honeysuckle ( Lonicera japonica) and Huangqi ( Astragalus membranaceus) Suppress SARS-CoV-2 Entry and COVID-19 Related Cytokine Storm in Vitro. Front Pharmacol 2022; 12:765553. [PMID: 35401158 PMCID: PMC8990830 DOI: 10.3389/fphar.2021.765553] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
COVID-19 is threatening human health worldwide but no effective treatment currently exists for this disease. Current therapeutic strategies focus on the inhibition of viral replication or using anti-inflammatory/immunomodulatory compounds to improve host immunity, but not both. Traditional Chinese medicine (TCM) compounds could be promising candidates due to their safety and minimal toxicity. In this study, we have developed a novel in silico bioinformatics workflow that integrates multiple databases to predict the use of honeysuckle (Lonicera japonica) and Huangqi (Astragalus membranaceus) as potential anti-SARS-CoV-2 agents. Using extracts from honeysuckle and Huangqi, these two herbs upregulated a group of microRNAs including let-7a, miR-148b, and miR-146a, which are critical to reduce the pathogenesis of SARS-CoV-2. Moreover, these herbs suppressed pro-inflammatory cytokines including IL-6 or TNF-α, which were both identified in the cytokine storm of acute respiratory distress syndrome, a major cause of COVID-19 death. Furthermore, both herbs partially inhibited the fusion of SARS-CoV-2 spike protein-transfected BHK-21 cells with the human lung cancer cell line Calu-3 that was expressing ACE2 receptors. These herbs inhibited SARS-CoV-2 Mpro activity, thereby alleviating viral entry as well as replication. In conclusion, our findings demonstrate that honeysuckle and Huangqi have the potential to be used as an inhibitor of SARS-CoV-2 virus entry that warrants further in vivo analysis and functional assessment of miRNAs to confirm their clinical importance. This fast-screening platform can also be applied to other drug discovery studies for other infectious diseases.
Collapse
Affiliation(s)
- Yuan-Chieh Yeh
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan.,Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ly Hien Doan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Zi-Yi Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,ASUS Intelligent Cloud Services, Taipei, Taiwan
| | - Li-Wei Chu
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-Hau Shi
- Department of Life Sciences and Institute of Genome Sciences, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Ray Lee
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan.,Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Tao Wu
- Division of Big Data, Phalanx Biotech Group, Hsinchu, Taiwan
| | - Chao-Hsiung Lin
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Life Sciences and Institute of Genome Sciences, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Aging and Health Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shu-Tuan Chiang
- Chuang Song Zong Pharmaceutical Co., Ltd. Ligang Plant, Pingtung, Taiwan
| | - Hui-Kang Liu
- National Research Institute of Chinese Medicine (NRICM), Ministry of Health and Welfare, Taipei, Taiwan.,Ph. D. Program in the Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan.,Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yueh-Hsin Ping
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Biophotonics, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center for Cancer Research, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Ying F Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung, Taipei, Taiwan.,Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung, Taipei, Taiwan.,Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
79
|
The COVID-19 Cell Signalling Problem: Spike, RAGE, PKC, p38, NFκB & IL-6 Hyper-Expression and the Human Ezrin Peptide, VIP, PKA-CREB Solution. IMMUNO 2022. [DOI: 10.3390/immuno2020017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
SARS-CoV-2 infection inhibits interferon expression, while hyper-activating innate-immune signalling and expression of pro-inflammatory cytokines. SARS-CoV-2 proteins: Spike, M and nsp6, nsp12 and nsp13 inhibit IFR3-mediated Type-1-interferon defence, but hyper-activate intracellular signalling, which leads to dysfunctional expression of pro-inflammatory cytokines, particularly IL-1β IL-6, IL-8, and TNFα. Ezrin, a sub-membrane adaptor-protein, organises multi-protein-complexes such as ezrin+NHERF1+NHE+CFTR, which control the density and location of ACE2 receptor expression on the luminal surface of airway-epithelial-cells, as well as determining susceptibility to SARS-CoV-2 infection. This protein complex is vital for lung-surfactant production for efficient gas-exchange. Ezrin also forms multi-protein-complexes that regulate signalling kinases; Ras, PKC, PI3K, and PKA. m-RAGE is a pattern-recognition-receptor of the innate immune system that is triggered by AGEs, which are chemically modified proteins common in the elderly and obese. m-RAGE forms multi-protein complexes with ezrin and TIRAP, a toll-like-receptor adaptor-protein. The main cause of COVID-19 is not viral infection but pro-inflammatory p38MAPK signalling mediated by TLRs and RAGE. In contrast, it appears that activated ezrin+PKA signalling results in the activation of transcription-factor CREB, which suppresses NFκB mediated pro-inflammatory cytokine expression. In addition, competition between ezrin and TIRAP to form multi-protein-complexes on membrane PIP2-lipid-rafts is a macromolecular-switch that changes the priority from innate immune activation programs to adaptive immune activation programs. Human Vasoactive Intestinal Peptide (VIP), and Human Ezrin Peptides (HEP-1 and RepG3) probably inhibit COVID-19 by activating the ezrin+PKA and ras>Raf>MEK>ERK>RSK>CREB>IL-10 signalling, which favours activation of adaptive immunity programs and inhibition of the dysfunctional innate-inflammation, the cause of COVID-19. HEP-1, RepG3, and VIP in individual human volunteers and in small clinical studies have been shown to be effective COVID-19 therapies, and seem to have a closely related mechanism of action.
Collapse
|
80
|
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has had an enormous impact on the world, affecting people's lifestyle, economy, and livelihood. Recently, with the development of vaccines, the number of infected cases has decreased. Many case reports have revealed that COVID-19 may induce other serious comorbidities such as anti-N-methyl-d-aspartate (anti-NMDA) receptor encephalitis. Anti-NMDA receptor encephalitis is an acute autoimmune disease that occurs more commonly in women than in men. To explore the association between COVID-19 and anti-NMDA receptor encephalitis, the microRNA (miRNA) biomarkers of COVID-19, anti-NMDA receptor encephalitis, and other related diseases from the literature are reviewed; then on the basis of these miRNA biomarkers, the relationship between COVID-19 and anti-NMDA receptor encephalitis is discussed. miRNAs are small non-coding RNAs that play important roles in cell differentiation, development, cell-cycle regulation, and apoptosis. miRNAs have been used as biological biomarkers for many diseases. The results in this study reveal that the relationship between anti-NMDA receptor encephalitis and COVID-19 infection or COVID-19 vaccination cannot be excluded; however, the risk that COVID-19 triggers the anti-NMDA receptor encephalitis is not high.
Collapse
Affiliation(s)
- Hsiuying Wang
- Institute of Statistics, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
81
|
Abbasi A, Kafil HS, Ozma MA, Sangtarash N, Sabahi S. Can food matrices be considered as a potential carrier for COVID-19? LE INFEZIONI IN MEDICINA 2022; 30:59-72. [PMID: 35350257 PMCID: PMC8929742 DOI: 10.53854/liim-3001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
Humanity is currently facing a life-threatening challenge from the infectious and epidemic disease SARS-CoV-2. To date, the various modes of transmission of the virus have not been fully elucidated. In this regard, there is a possibility of transmission of the virus through food products. The COVID-19 pandemic disease, like those associated with SARS and MERS, is transmitted mainly through the respiratory tract and airborne aerosol particles, but the presence of fragments of the genetic virus (RNA) in the feces of numerous patients proposes that their fecal-oral pathway may be expanded. In addition, people with gastrointestinal disorders such as atrophic gastritis and metaplasia may be susceptible to COVID-19 infection. Accordingly, food may act as a potential carrier of COVID-19 due to environmental or cross-contamination. According to the available evidence, the spread and possibility of transmission of COVID-19 contamination from humans to food products are possible. Beyond that, there is some evidence that some food sources of animal origin, such as pigs and rabbits, can be contaminated by COVID-19. Therefore, the transmission of the virus through some meat products may be conceivable. Due to the rapid release rate of COVID-19 and its stability in various milieus, especially food manufacturing circumstances, it may enter the matrix during different stages of traditional or industrial food processing. Therefore, preventive measures are recommended to be utilized in the food manufacturing sector. The present study explored the risk of different food matrices, including dairy products, bread, meat and meat products, vegetables, fruits, and processed foods, as potential carriers for the transmission of COVID-19.
Collapse
Affiliation(s)
- Amin Abbasi
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Asghari Ozma
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Sangtarash
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sahar Sabahi
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
82
|
Ning Q, Wu D, Wang X, Xi D, Chen T, Chen G, Wang H, Lu H, Wang M, Zhu L, Hu J, Liu T, Ma K, Han M, Luo X. The mechanism underlying extrapulmonary complications of the coronavirus disease 2019 and its therapeutic implication. Signal Transduct Target Ther 2022; 7:57. [PMID: 35197452 PMCID: PMC8863906 DOI: 10.1038/s41392-022-00907-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is a highly transmissible disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that poses a major threat to global public health. Although COVID-19 primarily affects the respiratory system, causing severe pneumonia and acute respiratory distress syndrome in severe cases, it can also result in multiple extrapulmonary complications. The pathogenesis of extrapulmonary damage in patients with COVID-19 is probably multifactorial, involving both the direct effects of SARS-CoV-2 and the indirect mechanisms associated with the host inflammatory response. Recognition of features and pathogenesis of extrapulmonary complications has clinical implications for identifying disease progression and designing therapeutic strategies. This review provides an overview of the extrapulmonary complications of COVID-19 from immunological and pathophysiologic perspectives and focuses on the pathogenesis and potential therapeutic targets for the management of COVID-19.
Collapse
Affiliation(s)
- Qin Ning
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Di Wu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Xi
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Chen
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guang Chen
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongwu Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Lu
- National Medical Center for Major Public Health Events, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Wang
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Zhu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjian Hu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Liu
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Ma
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meifang Han
- National Medical Center for Major Public Health Events, Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Luo
- National Medical Center for Major Public Health Events, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
83
|
Park SB, Irvin P, Hu Z, Khan M, Hu X, Zeng Q, Chen C, Xu M, Leek M, Zang R, Case JB, Zheng W, Ding S, Liang TJ. Targeting the Fusion Process of SARS-CoV-2 Infection by Small Molecule Inhibitors. mBio 2022; 13:e0323821. [PMID: 35012356 PMCID: PMC8749431 DOI: 10.1128/mbio.03238-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 12/31/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a serious threat to global public health, underscoring the urgency of developing effective therapies. Therapeutics and, more specifically, direct-acting antiviral development are still very much in their infancy. Here, we report that two hepatitis C virus (HCV) fusion inhibitors identified in our previous study, dichlorcyclizine and fluoxazolevir, broadly block human coronavirus entry into various cell types. Both compounds were effective against various human-pathogenic CoVs in multiple assays based on vesicular stomatitis virus (VSV) pseudotyped with the spike protein and spike-mediated syncytium formation. The antiviral effects were confirmed in SARS-CoV-2 infection systems. These compounds were equally effective against recently emerged variants, including the delta variant. Cross-linking experiments and structural modeling suggest that the compounds bind to a hydrophobic pocket near the fusion peptide of S protein, consistent with their potential mechanism of action as fusion inhibitors. In summary, these fusion inhibitors have broad-spectrum antiviral activities and may be promising leads for treatment of SARS-CoV-2, its variants, and other pathogenic CoVs. IMPORTANCE SARS-CoV-2 is an enveloped virus that requires membrane fusion for entry into host cells. Since the fusion process is relatively conserved among enveloped viruses, we tested our HCV fusion inhibitors, dichlorcyclizine and fluoxazolevir, against SARS-CoV-2. We performed in vitro assays and demonstrated their effective antiviral activity against SARS-CoV-2 and its variants. Cross-linking experiments and structural modeling suggest that the compounds bind to a hydrophobic pocket in spike protein to exert their inhibitory effect on the fusion step. These data suggest that both dichlorcyclizine and fluoxazolevir are promising candidates for further development as treatment for SARS-CoV-2.
Collapse
Affiliation(s)
- Seung Bum Park
- Liver Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | - Parker Irvin
- Liver Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | - Zongyi Hu
- Liver Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | - Mohsin Khan
- Liver Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | - Xin Hu
- Division of Pre-Clinical Innovations, NCATS, NIH, Rockville, Maryland, USA
| | - Qiru Zeng
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Catherine Chen
- Division of Pre-Clinical Innovations, NCATS, NIH, Rockville, Maryland, USA
| | - Miao Xu
- Division of Pre-Clinical Innovations, NCATS, NIH, Rockville, Maryland, USA
| | - Madeleine Leek
- Liver Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | - Ruochen Zang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - James Brett Case
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Wei Zheng
- Division of Pre-Clinical Innovations, NCATS, NIH, Rockville, Maryland, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - T. Jake Liang
- Liver Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| |
Collapse
|
84
|
de Carvalho Lima EN, Octaviano ALM, Piqueira JRC, Diaz RS, Justo JF. Coronavirus and Carbon Nanotubes: Seeking Immunological Relationships to Discover Immunotherapeutic Possibilities. Int J Nanomedicine 2022; 17:751-781. [PMID: 35241912 PMCID: PMC8887185 DOI: 10.2147/ijn.s341890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
Since December 2019, the world has faced an unprecedented pandemic crisis due to a new coronavirus disease, coronavirus disease-2019 (COVID-19), which has instigated intensive studies on prevention and treatment possibilities. Here, we investigate the relationships between the immune activation induced by three coronaviruses associated with recent outbreaks, with special attention to SARS-CoV-2, the causative agent of COVID-19, and the immune activation induced by carbon nanotubes (CNTs) to understand the points of convergence in immune induction and modulation. Evidence suggests that CNTs are among the most promising materials for use as immunotherapeutic agents. Therefore, this investigation explores new possibilities of effective immunotherapies for COVID-19. This study aimed to raise interest and knowledge about the use of CNTs as immunotherapeutic agents in coronavirus treatment. Thus, we summarize the most important immunological aspects of various coronavirus infections and describe key advances and challenges in using CNTs as immunotherapeutic agents against viral infections and the activation of the immune response induced by CNTs, which can shed light on the immunotherapeutic possibilities of CNTs.
Collapse
Affiliation(s)
- Elidamar Nunes de Carvalho Lima
- Telecommunication and Control Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
- Infectious Diseases Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
- Electronic Systems Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, SP, CEP 05508-010, Brazil
| | - Ana Luiza Moraes Octaviano
- Telecommunication and Control Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
| | - José Roberto Castilho Piqueira
- Telecommunication and Control Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
| | - Ricardo Sobhie Diaz
- Infectious Diseases Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - João Francisco Justo
- Electronic Systems Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, SP, CEP 05508-010, Brazil
| |
Collapse
|
85
|
Alnima T, Mulder MM, van Bussel BC, ten Cate H. COVID-19 Coagulopathy: From Pathogenesis to Treatment. Acta Haematol 2022; 145:282-296. [PMID: 35499460 PMCID: PMC9059042 DOI: 10.1159/000522498] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/01/2022] [Indexed: 12/25/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has emerged as a pandemic at the end of 2019 and continues to exert an unfavorable worldwide health impact on a large proportion of the population. A remarkable feature of COVID-19 is the precipitation of a hypercoagulable state, mainly in severe cases, leading to micro- and macrothrombosis, respiratory failure, and death. Despite the implementation of various therapeutic regimes, including anticoagulants, a large number of patients suffer from such serious complications. This review aims to describe the current knowledge on the pathophysiology of the coagulation mechanism in COVID-19. We describe the interplay between three important mediators of the disease and how this may lead to a hyperinflammatory and prothrombotic state that affects outcome, namely, the endothelium, the immune system, and the coagulation system. In line with the hypercoagulability state during COVID-19, we further review on the rare but severe vaccine-induced thrombotic thrombocytopenia. We also summarize and comment on available anticoagulant treatment options and include suggestions for some future treatment considerations for COVID-19 anticoagulation therapy.
Collapse
Affiliation(s)
- Teba Alnima
- Department of Internal Medicine, Vascular Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Mark M.G. Mulder
- Department of Intensive Care Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Bas C.T. van Bussel
- Department of Intensive Care Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Hugo ten Cate
- Department of Internal Medicine, Vascular Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
- *Hugo ten Cate,
| |
Collapse
|
86
|
Clark AL, Williams B. Recurrence of Pyoderma Gangrenosum Potentially Triggered by COVID-19 Vaccination. Cureus 2022; 14:e22625. [PMID: 35371773 PMCID: PMC8958150 DOI: 10.7759/cureus.22625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2022] [Indexed: 11/30/2022] Open
Abstract
Pyoderma gangrenosum (PG) is a rare inflammatory skin disease of unknown origin. As with other vaccines, COVID-19 vaccines have been associated with many cutaneous reactions. Although COVID-19 vaccination is crucial, it is important for dermatologists and other physicians to be aware of the possible cutaneous reactions that can occur following COVID-19 vaccination. In this report, we describe a 73-year-old woman with a personal history of PG who experienced a recurrence after receiving her second dose of the tozinameran vaccine. Although extremely rare, flares of other inflammatory dermatoses, including lichen planus, have been reported following COVID-19 vaccination. Here we discuss the overlap in pathogenesis of PG and COVID-19, proposing possible mechanisms behind this rare phenomenon.
Collapse
Affiliation(s)
- Abigale L Clark
- College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, Kansas City, USA
| | | |
Collapse
|
87
|
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
|
88
|
Liu W, Zeng Y, Li Y, Li N, Peng M, Cheng J, Tian B, Chen M. Exploring the Potential Targets and Mechanisms of Huang Lian Jie Du Decoction in the Treatment of Coronavirus Disease 2019 Based on Network Pharmacology. Int J Gen Med 2021; 14:9873-9885. [PMID: 34938107 PMCID: PMC8687521 DOI: 10.2147/ijgm.s337025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022] Open
Abstract
Background In December 2019, coronavirus disease 2019 (COVID-19) caused by a novel coronavirus (severe acute respiratory syndrome coronavirus 2, SARS-CoV-2; previously known as 2019-nCoV) emerged in Wuhan, China, and caused many infections and deaths. At present, there are no specific drugs for the etiology and treatment of COVID-19. A combination of traditional Chinese and western medicine is proposed to treat COVID-19, in which Huang Lian Jie Du decoction (HLJDD) is recommended for the treatment of COVID-19 in many provinces in China and has been widely used in the clinic. This study explored the potential targets of HLJDD in the treatment of COVID-19 based on network pharmacology. Methods First, the chemical composition and targets of HLJDD and COVID-19-related targets were obtained through the TCMSP, UniProt, GeneCards and OMIM databases. Second, HLJDD target and HLJDD-COVID-19 target networks were constructed via the STRING database and Cytoscape software. Finally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the HLJDD-COVID-19 targets was applied via the DAVID database. Results Our study identified a total of 67 active ingredients of HLJDD and 204 targets of HLJDD. A total of 502 COVID-19-related targets were obtained, of which 47 were intersecting targets of HLJDD and COVID-19. A total of 179 GO terms and 77 KEGG terms, including the TNF signaling pathway, NF-κB signaling pathway and HIF-1 signaling pathway, were identified. Conclusion The present study explored the potential targets and signaling pathways of HLJDD during the treatment of COVID-19, which may provide a basis for the research and development of drugs for the treatment of COVID-19.
Collapse
Affiliation(s)
- Wang Liu
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Yu Zeng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Yanda Li
- Department of Internal Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, People's Republic of China
| | - Nanhong Li
- Department of Pathology and Pathophysiology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Min Peng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Junfen Cheng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Binbin Tian
- Department of Critical Care Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, People's Republic of China
| | - Mingdi Chen
- Department of Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| |
Collapse
|
89
|
Vlasov I, Panteleeva A, Usenko T, Nikolaev M, Izumchenko A, Gavrilova E, Shlyk I, Miroshnikova V, Shadrina M, Polushin Y, Pchelina S, Slonimsky P. Transcriptomic Profiles Reveal Downregulation of Low-Density Lipoprotein Particle Receptor Pathway Activity in Patients Surviving Severe COVID-19. Cells 2021; 10:3495. [PMID: 34944005 PMCID: PMC8700658 DOI: 10.3390/cells10123495] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
To assess the biology of the lethal endpoint in patients with SARS-CoV-2 infection, we compared the transcriptional response to the virus in patients who survived or died during severe COVID-19. We applied gene expression profiling to generate transcriptional signatures for peripheral blood mononuclear cells (PBMCs) from patients with SARS-CoV-2 infection at the time when they were placed in the Intensive Care Unit of the Pavlov First State Medical University of St. Petersburg (Russia). Three different bioinformatics approaches to RNA-seq analysis identified a downregulation of three common pathways in survivors compared with nonsurvivors among patients with severe COVID-19, namely, low-density lipoprotein (LDL) particle receptor activity (GO:0005041), important for maintaining cholesterol homeostasis, leukocyte differentiation (GO:0002521), and cargo receptor activity (GO:0038024). Specifically, PBMCs from surviving patients were characterized by reduced expression of PPARG, CD36, STAB1, ITGAV, and ANXA2. Taken together, our findings suggest that LDL particle receptor pathway activity in patients with COVID-19 infection is associated with poor disease prognosis.
Collapse
Affiliation(s)
- Ivan Vlasov
- Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (I.V.); (M.S.)
| | - Alexandra Panteleeva
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint-Petersburg, Russia; (A.P.); (T.U.); (M.N.); (E.G.); (I.S.); (V.M.); (Y.P.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Saint-Petersburg, Russia;
| | - Tatiana Usenko
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint-Petersburg, Russia; (A.P.); (T.U.); (M.N.); (E.G.); (I.S.); (V.M.); (Y.P.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Saint-Petersburg, Russia;
| | - Mikhael Nikolaev
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint-Petersburg, Russia; (A.P.); (T.U.); (M.N.); (E.G.); (I.S.); (V.M.); (Y.P.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Saint-Petersburg, Russia;
| | - Artem Izumchenko
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Saint-Petersburg, Russia;
| | - Elena Gavrilova
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint-Petersburg, Russia; (A.P.); (T.U.); (M.N.); (E.G.); (I.S.); (V.M.); (Y.P.); (S.P.)
| | - Irina Shlyk
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint-Petersburg, Russia; (A.P.); (T.U.); (M.N.); (E.G.); (I.S.); (V.M.); (Y.P.); (S.P.)
| | - Valentina Miroshnikova
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint-Petersburg, Russia; (A.P.); (T.U.); (M.N.); (E.G.); (I.S.); (V.M.); (Y.P.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Saint-Petersburg, Russia;
| | - Maria Shadrina
- Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (I.V.); (M.S.)
| | - Yurii Polushin
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint-Petersburg, Russia; (A.P.); (T.U.); (M.N.); (E.G.); (I.S.); (V.M.); (Y.P.); (S.P.)
| | - Sofya Pchelina
- Pavlov First Saint-Petersburg State Medical University, 197022 Saint-Petersburg, Russia; (A.P.); (T.U.); (M.N.); (E.G.); (I.S.); (V.M.); (Y.P.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Saint-Petersburg, Russia;
- Kurchatov Genome Center—PNPI, 188300 Saint-Petersburg, Russia
| | - Petr Slonimsky
- Institute of Molecular Genetics of National Research Center “Kurchatov Institute”, 123182 Moscow, Russia; (I.V.); (M.S.)
| |
Collapse
|
90
|
Lin Z, Niu J, Xu Y, Qin L, Ding J, Zhou L. Clinical efficacy and adverse events of baricitinib treatment for coronavirus disease-2019 (COVID-19): A systematic review and meta-analysis. J Med Virol 2021; 94:1523-1534. [PMID: 34846766 PMCID: PMC9015594 DOI: 10.1002/jmv.27482] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/08/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022]
Abstract
The benefits of baricitinib in coronavirus disease-2019 are inadequately defined. We performed a systematic review and meta-analysis of studies of baricitinib to determine its clinical efficacy and adverse events in patients with COVID-19. Databases were searched from their inception to September 5, 2021. The primary outcome was the coefficient of mortality. We also compared secondary indicators and adverse events between baricitinib treatment and placebo or other treatments. Twelve studies of 3564 patients were included and assessed qualitatively (modified Jadad and Newcastle-Ottawa Scale scores). Baricitinib effectively improved the mortality rate (relative risk of mortality = 0.56; 95% confidence interval: 0.46-0.69; p < 0.001; I2 = 2%), and this result was unchanged by subgroup analysis. Baricitinib improved intensive care unit admission, the requirement for invasive mechanical ventilation, and improved the oxygenation index. Data from these studies also showed that baricitinib slightly reduced the risk of adverse events. Regarding the choice of the drug dosage of baricitinib, the high-dose group appeared to have additional benefits for clinical efficacy. Our study shows that baricitinib may be a promising, safe, and effective anti-severe acute respiratory syndrome-coronavirus-2 drug candidate, with the advantages of low cost, easy production, and convenient storage.
Collapse
Affiliation(s)
- Zhiwei Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianyi Niu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifan Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijie Qin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiabin Ding
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Luqian Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
91
|
Dewanjee S, Kandimalla R, Kalra RS, Valupadas C, Vallamkondu J, Kolli V, Dey Ray S, Reddy AP, Reddy PH. COVID-19 and Rheumatoid Arthritis Crosstalk: Emerging Association, Therapeutic Options and Challenges. Cells 2021; 10:3291. [PMID: 34943795 PMCID: PMC8699554 DOI: 10.3390/cells10123291] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 02/07/2023] Open
Abstract
Hyperactivation of immune responses resulting in excessive release of pro-inflammatory mediators in alveoli/lung structures is the principal pathological feature of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The cytokine hyperactivation in COVID-19 appears to be similar to those seen in rheumatoid arthritis (RA), an autoimmune disease. Emerging evidence conferred the severity and risk of COVID-19 to RA patients. Amid the evidence of musculoskeletal manifestations involving immune-inflammation-dependent mechanisms and cases of arthralgia and/or myalgia in COVID-19, crosstalk between COVID-19 and RA is often debated. The present article sheds light on the pathological crosstalk between COVID-19 and RA, the risk of RA patients in acquiring SARS-CoV-2 infection, and the aspects of SARS-CoV-2 infection in RA development. We also conferred whether RA can exacerbate COVID-19 outcomes based on available clinical readouts. The mechanistic overlapping in immune-inflammatory features in both COVID-19 and RA was discussed. We showed the emerging links of angiotensin-converting enzyme (ACE)-dependent and macrophage-mediated pathways in both diseases. Moreover, a detailed review of immediate challenges and key recommendations for anti-rheumatic drugs in the COVID-19 setting was presented for better clinical monitoring and management of RA patients. Taken together, the present article summarizes available knowledge on the emerging COVID-19 and RA crosstalk and their mechanistic overlaps, challenges, and therapeutic options.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur Unversity, Kolkata 700032, India;
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Technology, Uppal Road, Tarnaka, Hyderabad 50000, India;
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, India
| | - Rajkumar Singh Kalra
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Higashi 1-1-1, Tsukuba 305-8565, Japan;
| | - Chandrasekhar Valupadas
- Department of Medicine, Mahatma Gandhi Memorial Hospital, Warangal 506007, India;
- Department of Medicine, Kakatiya Medical College Superspeciality Hospital, Warangal 506007, India
| | | | - Viswakalyan Kolli
- Department of Biochemistry, GITAM Institute of Medical Sciences and Research, Visakhapatnam 530045, India;
| | - Sarbani Dey Ray
- Department of Pharmaceutical Sciences, Assam University, Silchar 788011, India;
| | - Arubala P. Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA;
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Departments of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
92
|
Bliek-Bueno K, Mucherino S, Poblador-Plou B, González-Rubio F, Aza-Pascual-Salcedo M, Orlando V, Clerencia-Sierra M, Ioakeim-Skoufa I, Coscioni E, Carmona-Pírez J, Perrella A, Trama U, Prados-Torres A, Menditto E, Gimeno-Miguel A. Baseline Drug Treatments as Indicators of Increased Risk of COVID-19 Mortality in Spain and Italy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:11786. [PMID: 34831541 PMCID: PMC8623536 DOI: 10.3390/ijerph182211786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 02/05/2023]
Abstract
This study aims to identify baseline medications that, as a proxy for the diseases they are dispensed for, are associated with increased risk of mortality in COVID-19 patients from two regions in Spain and Italy using real-world data. We conducted a cross-country, retrospective, observational study including 8570 individuals from both regions with confirmed SARS-CoV-2 infection between 4 March and 17 April 2020, and followed them for a minimum of 30 days to allow sufficient time for the studied event, in this case death, to occur. Baseline demographic variables and all drugs dispensed in community pharmacies three months prior to infection were extracted from the PRECOVID Study cohort (Aragon, Spain) and the Campania Region Database (Campania, Italy) and analyzed using logistic regression models. Results show that the presence at baseline of potassium-sparing agents, antipsychotics, vasodilators, high-ceiling diuretics, antithrombotic agents, vitamin B12, folic acid, and antiepileptics were systematically associated with mortality in COVID-19 patients from both countries. Treatments for chronic cardiovascular and metabolic diseases, systemic inflammation, and processes with increased risk of thrombosis as proxies for the conditions they are intended for can serve as timely indicators of an increased likelihood of mortality after the infection, and the assessment of pharmacological profiles can be an additional approach to the identification of at-risk individuals in clinical practice.
Collapse
Affiliation(s)
- Kevin Bliek-Bueno
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Teaching Unit of Preventive Medicine and Public Health, Miguel Servet University Hospital, 50009 Zaragoza, Spain
| | - Sara Mucherino
- Centro Interdipartimentale di Ricerca in Farmacoeconomia e Farmacoutilizzazione (CIRFF), Center of Drug Utilization and Pharmacoeconomics, Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (V.O.); (E.M.)
| | - Beatriz Poblador-Plou
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Health Services Research on Chronic Patients Network (REDISSEC), Institute of Health Carlos III (ISCIII), 28222 Madrid, Spain
| | - Francisca González-Rubio
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Health Services Research on Chronic Patients Network (REDISSEC), Institute of Health Carlos III (ISCIII), 28222 Madrid, Spain
- Delicias-Sur Primary Care Health Centre, Aragon Health Service (SALUD), 50009 Zaragoza, Spain
- Drug Utilization Work Group, Spanish Society of Family and Community Medicine (semFYC), 08009 Barcelona, Spain
| | - Mercedes Aza-Pascual-Salcedo
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Health Services Research on Chronic Patients Network (REDISSEC), Institute of Health Carlos III (ISCIII), 28222 Madrid, Spain
- Primary Care Pharmacy Service Zaragoza III, Aragon Health Service (SALUD), 50017 Zaragoza, Spain
| | - Valentina Orlando
- Centro Interdipartimentale di Ricerca in Farmacoeconomia e Farmacoutilizzazione (CIRFF), Center of Drug Utilization and Pharmacoeconomics, Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (V.O.); (E.M.)
| | - Mercedes Clerencia-Sierra
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Health Services Research on Chronic Patients Network (REDISSEC), Institute of Health Carlos III (ISCIII), 28222 Madrid, Spain
- Aragon Health Service (SALUD), Miguel Servet University Hospital, 50009 Zaragoza, Spain
| | - Ignatios Ioakeim-Skoufa
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Drug Utilization Work Group, Spanish Society of Family and Community Medicine (semFYC), 08009 Barcelona, Spain
- WHO Collaborating Centre for Drug Statistics Methodology, Norwegian Institute of Public Health, 0213 Oslo, Norway
- Department of Drug Statistics, Division of Health Data and Digitalisation, Norwegian Institute of Public Health, 0213 Oslo, Norway
| | - Enrico Coscioni
- Division of Cardiac Surgery, AOU San Giovanni di Dio e Ruggi d’Aragona, 84131 Salerno, Italy;
| | - Jonás Carmona-Pírez
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Health Services Research on Chronic Patients Network (REDISSEC), Institute of Health Carlos III (ISCIII), 28222 Madrid, Spain
- Delicias-Sur Primary Care Health Centre, Aragon Health Service (SALUD), 50009 Zaragoza, Spain
| | - Alessandro Perrella
- Infectious Disease of Healthcare Direction, AORN Antonio Cardarelli, 80131 Naples, Italy;
| | - Ugo Trama
- Regional Pharmaceutical Unit, Campania Region, 80143 Naples, Italy;
| | - Alexandra Prados-Torres
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Health Services Research on Chronic Patients Network (REDISSEC), Institute of Health Carlos III (ISCIII), 28222 Madrid, Spain
| | - Enrica Menditto
- Centro Interdipartimentale di Ricerca in Farmacoeconomia e Farmacoutilizzazione (CIRFF), Center of Drug Utilization and Pharmacoeconomics, Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (V.O.); (E.M.)
| | - Antonio Gimeno-Miguel
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), IIS Aragón, Miguel Servet University Hospital, 50009 Zaragoza, Spain; (K.B.-B.); (B.P.-P.); (F.G.-R.); (M.A.-P.-S.); (M.C.-S.); (I.I.-S.); (J.C.-P.); (A.P.-T.)
- Health Services Research on Chronic Patients Network (REDISSEC), Institute of Health Carlos III (ISCIII), 28222 Madrid, Spain
| |
Collapse
|
93
|
Neurological Manifestations and Outcomes in a Retrospective Cohort of Mexican Inpatients with SARS-CoV-2 Pneumonia: Design of a Risk Profile. Healthcare (Basel) 2021; 9:healthcare9111501. [PMID: 34828547 PMCID: PMC8620259 DOI: 10.3390/healthcare9111501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 01/08/2023] Open
Abstract
We analyzed the neurological manifestations in Mexican patients hospitalized with pneumonia due to COVID-19 and investigated the association between demographic, clinical, and biochemical variables and outcomes, including death. A retrospective, analytical study was conducted using the electronic records of patients hospitalized between 1 April 2020 and 30 September 2020. Records of 1040 patients were analyzed: 31.25% died and 79.42% had neurological symptoms, including headache (80.62%), anosmia (32.20%), ageusia (31.96%), myopathy (28.08%), disorientation (14.89%), encephalopathy (12.22%), neuropathy (5.4%), stroke (1.3%), seizures (1.3%), cerebral hemorrhage (1.08%), encephalitis (0.84%), central venous thrombosis (0.36%), and subarachnoid hemorrhage (0.24%). Patients also had comorbidities, such as hypertension (42.30%), diabetes mellitus (38.74%), obesity (61.34%), chronic obstructive pulmonary disease (3.17%), and asthma (2.01%). Factors associated with neurological symptoms were dyspnea, chronic obstructive pulmonary disease, advanced respiratory support, prolonged hospitalization, and worsening fibrinogen levels. Factors associated with death were older age, advanced respiratory support, amine management, chronic obstructive pulmonary disease, intensive care unit management, dyspnea, disorientation, encephalopathy, hypertension, neuropathy, diabetes, male sex, three or more neurological symptoms, and obesity grade 3. In this study we designed a profile to help predict patients at higher risk of developing neurological complications and death following COVID-19 infection.
Collapse
|
94
|
Astore C, Zhou H, Jacob J, Skolnick J. Prediction of severe adverse events, modes of action and drug treatments for COVID-19's complications. Sci Rep 2021; 11:20864. [PMID: 34675303 PMCID: PMC8531388 DOI: 10.1038/s41598-021-00368-6] [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: 07/02/2021] [Accepted: 10/06/2021] [Indexed: 01/08/2023] Open
Abstract
Following SARS-CoV-2 infection, some COVID-19 patients experience severe host driven adverse events. To treat these complications, their underlying etiology and drug treatments must be identified. Thus, a novel AI methodology MOATAI-VIR, which predicts disease-protein-pathway relationships and repurposed FDA-approved drugs to treat COVID-19's clinical manifestations was developed. SARS-CoV-2 interacting human proteins and GWAS identified respiratory failure genes provide the input from which the mode-of-action (MOA) proteins/pathways of the resulting disease comorbidities are predicted. These comorbidities are then mapped to their clinical manifestations. To assess each manifestation's molecular basis, their prioritized shared proteins were subject to global pathway analysis. Next, the molecular features associated with hallmark COVID-19 phenotypes, e.g. unusual neurological symptoms, cytokine storms, and blood clots were explored. In practice, 24/26 of the major clinical manifestations are successfully predicted. Three major uncharacterized manifestation categories including neoplasms are also found. The prevalence of neoplasms suggests that SARS-CoV-2 might be an oncovirus due to shared molecular mechanisms between oncogenesis and viral replication. Then, repurposed FDA-approved drugs that might treat COVID-19's clinical manifestations are predicted by virtual ligand screening of the most frequent comorbid protein targets. These drugs might help treat both COVID-19's severe adverse events and lesser ones such as loss of taste/smell.
Collapse
Affiliation(s)
- Courtney Astore
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA
| | - Hongyi Zhou
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA
| | - Joshy Jacob
- Emory Vaccine Center, Emory University, Atlanta, GA, 30329, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Microbiology and Immunology, Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, 30329, USA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA, 30332, USA.
| |
Collapse
|
95
|
Parums DV. Editorial: Multisystem Inflammatory Syndrome in Adults (MIS-A) and the Spectrum of COVID-19. Med Sci Monit 2021; 27:e935005. [PMID: 34629462 PMCID: PMC8518511 DOI: 10.12659/msm.935005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recent studies on the pathogenesis and clinical spectrum of human disease following infection with the new human pathogen, SARS-CoV-2, have identified the varied presentations and sequelae of COVID-19. Acute ‘cytokine storm’ in severe COVID-19 results in multiorgan damage due to vascular hyperpermeability, edema, and hypercoagulation. The long-term consequences of infection from SARS-CoV-2 include long COVID. or post-COVID syndrome, and multisystem inflammatory syndrome in children (MIS-C). Several case reports of multisystem inflammatory syndrome in adults (MIS-A) have shown the presentation at more than four weeks after initial infection with SARS-CoV-2 in adults more than 21 years of age. In September 2021, a published systematic review of the literature identified 221 patients with MIS-A, representing the most comprehensive clinical study to date. MIS-A occurs in the post-acute COVID-19 period. The pathogenesis may involve a dysregulated antibody-mediated immune response, similar to MIS-C. Therefore, patients with MIS-A may respond to supportive therapies that control hyperinflammation. This Editorial aims to describe MIS-A and discuss COVID-19 as a spectrum of hyperinflammatory disease in terms of severity, extent, duration, and patient age.
Collapse
Affiliation(s)
- Dinah V Parums
- Science Editor, Medical Science Monitor, International Scientific Information, Inc., Melville, NY, USA
| |
Collapse
|
96
|
Tan LY, Komarasamy TV, RMT Balasubramaniam V. Hyperinflammatory Immune Response and COVID-19: A Double Edged Sword. Front Immunol 2021; 12:742941. [PMID: 34659238 PMCID: PMC8515020 DOI: 10.3389/fimmu.2021.742941] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 09/10/2021] [Indexed: 12/14/2022] Open
Abstract
The coronavirus disease-19 (COVID-19) elicited by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused devastating health, economic and social impact worldwide. Its clinical spectrum ranges from asymptomatic to respiratory failure and multi-organ failure or death. The pathogenesis of SARS-CoV-2 infection is attributed to a complex interplay between virus and host immune response. It involves activation of multiple inflammatory pathways leading to hyperinflammation and cytokine storm, resulting in tissue damage, acute respiratory distress syndrome (ARDS) and multi-organ failure. Accumulating evidence has raised concern over the long-term health effects of COVID-19. Importantly, the neuroinvasive potential of SARS-CoV-2 may have devastating consequences in the brain. This review provides a conceptual framework on how the virus tricks the host immune system to induce infection and cause severe disease. We also explore the key differences between mild and severe COVID-19 and its short- and long-term effects, particularly on the human brain.
Collapse
Affiliation(s)
- Li Yin Tan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
- Greenslopes Private Hospital, Greenslopes, QLD, Australia
| | - Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Vinod RMT Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
97
|
Castle RD, Williams MA, Bushell WC, Rindfleisch JA, Peterson CT, Marzolf J, Brouwer K, Mills PJ. Implications for Systemic Approaches to COVID-19: Effect Sizes of Remdesivir, Tocilizumab, Melatonin, Vitamin D3, and Meditation. J Inflamm Res 2021; 14:4859-4876. [PMID: 34588793 PMCID: PMC8473718 DOI: 10.2147/jir.s323356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/28/2021] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION COVID-19 poses a chronic threat to inflammatory systems, reinforcing the need for efficient anti-inflammatory strategies. The purpose of this review and analysis was to determine the efficacy of various interventions upon the inflammatory markers most affected by COVID-19. The focus was on the markers associated with COVID-19, not the etiology of the virus itself. METHODS Based on 27 reviewed papers, information was extracted on the effects of COVID-19 upon inflammatory markers, then the effects of standard treatments (Remdesivir, Tocilizumab) and adjunctive interventions (vitamin D3, melatonin, and meditation) were extracted for those markers. These data were used to approximate effect sizes for the disease or interventions via standardized mean differences (SMD). RESULTS The data that were available indicated that adjunctive interventions affected 68.4% of the inflammatory markers impacted by COVID-19, while standard pharmaceutical medication affected 26.3%. DISCUSSION Nonstandard adjunctive care appeared to have comparable or superior effects in comparison to Remdesivir and Tocilizumab on the inflammatory markers most impacted by COVID-19. Alongside standards of care, melatonin, vitamin D3, and meditation should be considered for treatment of SARS-COV-2 infection and COVID-19 disease.
Collapse
Affiliation(s)
- Ryan D Castle
- Science Division, Whole Health Institute, Bentonville, AR, USA
| | - Michelle A Williams
- Harvard T.H. Chan School of Public Health, Department of Global Health and Population, Harvard University, Boston, MA, USA
| | | | - J Adam Rindfleisch
- Education Department, Whole Health School of Medicine and Health Sciences, Bentonville, AR, USA
| | - Christine Tara Peterson
- Center of Excellence for Research and Training in Integrative Health, Department of Family Medicine, School of Medicine, University of California, San Diego, CA, USA
| | - James Marzolf
- Health Sector Finance & Policy, Whole Health Institute, Bentonville, AR, USA
| | - Kimberly Brouwer
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, CA, USA
| | - Paul J Mills
- Herbert Wertheim School of Public Health and Human Longevity Science, Center of Excellence for Research and Training in Integrative Health, University of California, San Diego, CA, USA
| |
Collapse
|
98
|
Nazerian Y, Vakili K, Ebrahimi A, Niknejad H. Developing Cytokine Storm-Sensitive Therapeutic Strategy in COVID-19 Using 8P9R Chimeric Peptide and Soluble ACE2. Front Cell Dev Biol 2021; 9:717587. [PMID: 34540833 PMCID: PMC8446510 DOI: 10.3389/fcell.2021.717587] [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: 05/31/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
Currently, the COVID-19 pandemic is an international challenge, largely due to lack of effective therapies. Pharmacotherapy has not yet been able to find a definitive treatment for COVID-19. Since SARS-CoV-2 affects several organs, treatment strategies that target the virus in a wider range are expected to be ultimately more successful. To this end, a two-step treatment strategy has been presented. In the first phase of the disease, when the patient is newly infected with the virus and the cytokine storm has not yet been developed, a chimeric peptide is used to inhibit virus entry into the host cell cytosol (by inhibiting endosomal pH acidification) and viral replication. After the virus entry and decrease of angiotensin converting enzyme 2 (ACE2) level, some people are unable to properly compensate for the ACE2 pathway and progress toward the cytokine storm. In the beginning of the cytokine storm, sACE2 protein is very effective in regulating the immune system toward the anti-inflammatory pathway, including M2 macrophages. Hence, the genes of 8P9R chimeric peptide and sACE2 would be inserted in an episomal vector with a separate promoter for each gene: the chimeric peptide gene promoter is a CMV promoter, while the sACE2 gene promoter is a NF-κB-sensitive promoter. The NF-κB-sensitive promoter induces the expression of sACE2 gene soon after elevation of NF-κB which is the main transcription factor of inflammatory genes. Thus, as the expression of inflammatory cytokines increases, the expression of sACE2 increases simultaneously. In this condition, sACE2 can prevent the cytokine storm by inhibiting the pro-inflammatory pathways. To deliver the designed vector to the target cells, mesenchymal stem cell-derived (MSC-derived) exosome-liposome hybrids are used. Herein, the strategy can be considered as a personalized clinical therapy for COVID-19, that can prevent morbidity and mortality in the future.
Collapse
Affiliation(s)
- Yasaman Nazerian
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ebrahimi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
99
|
Pushparaj PN, Abdulkareem AA, Naseer MI. Identification of Novel Gene Signatures using Next-Generation Sequencing Data from COVID-19 Infection Models: Focus on Neuro-COVID and Potential Therapeutics. Front Pharmacol 2021; 12:688227. [PMID: 34531741 PMCID: PMC8438179 DOI: 10.3389/fphar.2021.688227] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 is the causative agent for coronavirus disease-19 (COVID-19) and belongs to the family Coronaviridae that causes sickness varying from the common cold to more severe illnesses such as severe acute respiratory syndrome, sudden stroke, neurological complications (Neuro-COVID), multiple organ failure, and mortality in some patients. The gene expression profiles of COVID-19 infection models can be used to decipher potential therapeutics for COVID-19 and related pathologies, such as Neuro-COVID. Here, we used the raw RNA-seq reads (Single-End) in quadruplicates derived using Illumina Next Seq 500 from SARS-CoV-infected primary human bronchial epithelium (NHBE) and mock-treated NHBE cells obtained from the Gene Expression Omnibus (GEO) (GSE147507), and the quality control (QC) was evaluated using the CLC Genomics Workbench 20.0 (Qiagen, United States) before the RNA-seq analysis using BioJupies web tool and iPathwayGuide for gene ontologies (GO), pathways, upstream regulator genes, small molecules, and natural products. Additionally, single-cell transcriptomics data (GSE163005) of meta clusters of immune cells from the cerebrospinal fluid (CSF), such as T-cells/natural killer cells (NK) (TcMeta), dendritic cells (DCMeta), and monocytes/granulocyte (monoMeta) cell types for comparison, namely, Neuro-COVID versus idiopathic intracranial hypertension (IIH), were analyzed using iPathwayGuide. L1000 fireworks display (L1000FWD) and L1000 characteristic direction signature search engine (L1000 CDS2) web tools were used to uncover the small molecules that could potentially reverse the COVID-19 and Neuro-COVID-associated gene signatures. We uncovered small molecules such as camptothecin, importazole, and withaferin A, which can potentially reverse COVID-19 associated gene signatures. In addition, withaferin A, trichostatin A, narciclasine, camptothecin, and JQ1 have the potential to reverse Neuro-COVID gene signatures. Furthermore, the gene set enrichment analysis (GSEA) preranked method and Metascape web tool were used to decipher and annotate the gene signatures that were potentially reversed by these small molecules. In conclusion, our study unravels a rapid approach for applying next-generation knowledge discovery (NGKD) platforms to discover small molecules with therapeutic potential against COVID-19 and its related disease pathologies.
Collapse
Affiliation(s)
- Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Angham Abdulrahman Abdulkareem
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Imran Naseer
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
100
|
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
|