151
|
Guest PC, Abbasifard M, Jamialahmadi T, Majeed M, Kesharwani P, Sahebkar A. Multiplex Testing of Oxidative-Reductive Pathway in Patients with COVID-19. Methods Mol Biol 2022; 2511:333-344. [PMID: 35838972 DOI: 10.1007/978-1-0716-2395-4_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Infection with SARS-CoV-2, the causative agent of COVID-19, causes numerous cellular dysfunctions. The virus enters the host cells and hijacks the cell machinery for its replication, resulting in disturbances of the oxidative, reductive balance, increased production of damaging reactive oxygen species (ROS), and mitochondrial dysfunction. This damaging cycle can make cells less resistant to infection and make the host more likely to experience a severe disease course. Treatment with antioxidants has been tested as a potential approach to reduce the effects of this disorder. Here, we present a protocol to assess the impact of treatment with a mixture of curcuminoids on physiological and molecular biomarkers, focusing on determining total antioxidant capacity. We used a cohort of diabetes patients with an imbalance in redox mechanisms as such patients are more likely to become severely ill from COVID-19 than healthy persons.
Collapse
Affiliation(s)
- Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Tannaz Jamialahmadi
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Amirhossein Sahebkar
- Applied Biomeical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Medicine, The University of Western Australia, Perth, WA, Australia.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
152
|
Dauby N, Flamand V. From maternal breath to infant's cells: Impact of maternal respiratory infections on infants 'immune responses. Front Pediatr 2022; 10:1046100. [PMID: 36419921 PMCID: PMC9676445 DOI: 10.3389/fped.2022.1046100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
In utero exposure to maternally-derived antigens following chronic infection is associated with modulation of infants 'immune response, differential susceptibility to post-natal infections and immune response toward vaccines. The maternal environment, both internal (microbiota) and external (exposure to environmental microbes) also modulates infant's immune response but also the clinical phenotype after birth. Vertical transmission of ubiquitous respiratory pathogens such as influenza and COVID-19 is uncommon. Evidence suggest that in utero exposure to maternal influenza and SARS-CoV-2 infections may have a significant impact on the developing immune system with activation of both innate and adaptive responses, possibly related to placental inflammation. Here in, we review how maternal respiratory infections, associated with airway, systemic and placental inflammation but also changes in maternal microbiota might impact infant's immune responses after birth. The clinical impact of immune modifications observed following maternal respiratory infections remains unexplored. Given the high frequencies of respiratory infections during pregnancy (COVID-19, influenza but also RSV and HMPV), the impact on global child health could be important.
Collapse
Affiliation(s)
- Nicolas Dauby
- Institute for Medical Immunology, ULB Center for Research in Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Department of Infectious Diseases, CHU Saint-Pierre, Brussels, Belgium.,School of Public Health, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Véronique Flamand
- Institute for Medical Immunology, ULB Center for Research in Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
153
|
Fazio S, Bellavite P, Zanolin E, McCullough PA, Pandolfi S, Affuso F. Retrospective Study of Outcomes and Hospitalization Rates of Patients in Italy with a Confirmed Diagnosis of Early COVID-19 and Treated at Home Within 3 Days or After 3 Days of Symptom Onset with Prescribed and Non-Prescribed Treatments Between November 2020 and August 2021. Med Sci Monit 2021; 27:e935379. [PMID: 34966165 PMCID: PMC8725339 DOI: 10.12659/msm.935379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/25/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND This retrospective study aimed to investigate outcomes and hospitalization rates in patients with a confirmed diagnosis of early COVID-19 treated at home with prescribed and non-prescribed treatments. MATERIAL AND METHODS The medical records of a cohort of 158 Italian patients with early COVID-19 treated at home were analyzed. Treatments consisted of indomethacin, low-dose aspirin, omeprazole, and a flavonoid-based food supplement, plus azithromycin, low-molecular-weight heparin, and betamethasone as needed. The association of treatment timeliness and of clinical variables with the duration of symptoms and with the risk of hospitalization was evaluated by logistic regression. RESULTS Patients were divided into 2 groups: group 1 (n=85) was treated at the earliest possible time (<72 h from onset of symptoms), and group 2 (n=73) was treated >72 h after the onset of symptoms. Clinical severity at the beginning of treatment was similar in the 2 groups. In group 1, symptom duration was shorter than in group 2 (median 6.0 days vs 13.0 days, P<0.001) and no hospitalizations occurred, compared with 19.18% hospitalizations in group 2. One patient in group 1 developed chest X-ray alterations and 2 patients experienced an increase in D-dimer levels, compared with 30 and 22 patients, respectively, in group 2. The main factor determining the duration of symptoms and the risk of hospitalization was the delay in starting therapy (P<0.001). CONCLUSIONS This real-world study of patients in the community showed that early diagnosis and early supportive patient management reduced the severity of COVID-19 and reduced the rate of hospitalization.
Collapse
Affiliation(s)
- Serafino Fazio
- Retired Professor of Internal Medicine, Medical School University Federico II, Naples, Italy
| | - Paolo Bellavite
- Physiopathology Chair, Homeopathic Medical School of Verona, Verona, Italy
| | - Elisabetta Zanolin
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Sergio Pandolfi
- Department of Neurosurgery, Villa Mafalda Clinics, Rome, Italy
| | | |
Collapse
|
154
|
Hasankhani A, Bahrami A, Sheybani N, Aria B, Hemati B, Fatehi F, Ghaem Maghami Farahani H, Javanmard G, Rezaee M, Kastelic JP, Barkema HW. Differential Co-Expression Network Analysis Reveals Key Hub-High Traffic Genes as Potential Therapeutic Targets for COVID-19 Pandemic. Front Immunol 2021; 12:789317. [PMID: 34975885 PMCID: PMC8714803 DOI: 10.3389/fimmu.2021.789317] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background The recent emergence of COVID-19, rapid worldwide spread, and incomplete knowledge of molecular mechanisms underlying SARS-CoV-2 infection have limited development of therapeutic strategies. Our objective was to systematically investigate molecular regulatory mechanisms of COVID-19, using a combination of high throughput RNA-sequencing-based transcriptomics and systems biology approaches. Methods RNA-Seq data from peripheral blood mononuclear cells (PBMCs) of healthy persons, mild and severe 17 COVID-19 patients were analyzed to generate a gene expression matrix. Weighted gene co-expression network analysis (WGCNA) was used to identify co-expression modules in healthy samples as a reference set. For differential co-expression network analysis, module preservation and module-trait relationships approaches were used to identify key modules. Then, protein-protein interaction (PPI) networks, based on co-expressed hub genes, were constructed to identify hub genes/TFs with the highest information transfer (hub-high traffic genes) within candidate modules. Results Based on differential co-expression network analysis, connectivity patterns and network density, 72% (15 of 21) of modules identified in healthy samples were altered by SARS-CoV-2 infection. Therefore, SARS-CoV-2 caused systemic perturbations in host biological gene networks. In functional enrichment analysis, among 15 non-preserved modules and two significant highly-correlated modules (identified by MTRs), 9 modules were directly related to the host immune response and COVID-19 immunopathogenesis. Intriguingly, systemic investigation of SARS-CoV-2 infection identified signaling pathways and key genes/proteins associated with COVID-19's main hallmarks, e.g., cytokine storm, respiratory distress syndrome (ARDS), acute lung injury (ALI), lymphopenia, coagulation disorders, thrombosis, and pregnancy complications, as well as comorbidities associated with COVID-19, e.g., asthma, diabetic complications, cardiovascular diseases (CVDs), liver disorders and acute kidney injury (AKI). Topological analysis with betweenness centrality (BC) identified 290 hub-high traffic genes, central in both co-expression and PPI networks. We also identified several transcriptional regulatory factors, including NFKB1, HIF1A, AHR, and TP53, with important immunoregulatory roles in SARS-CoV-2 infection. Moreover, several hub-high traffic genes, including IL6, IL1B, IL10, TNF, SOCS1, SOCS3, ICAM1, PTEN, RHOA, GDI2, SUMO1, CASP1, IRAK3, HSPA5, ADRB2, PRF1, GZMB, OASL, CCL5, HSP90AA1, HSPD1, IFNG, MAPK1, RAB5A, and TNFRSF1A had the highest rates of information transfer in 9 candidate modules and central roles in COVID-19 immunopathogenesis. Conclusion This study provides comprehensive information on molecular mechanisms of SARS-CoV-2-host interactions and identifies several hub-high traffic genes as promising therapeutic targets for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Behzad Aria
- Department of Physical Education and Sports Science, School of Psychology and Educational Sciences, Yazd University, Yazd, Iran
| | - Behzad Hemati
- Biotechnology Research Center, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Farhang Fatehi
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | | | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Mahsa Rezaee
- Department of Medical Mycology, School of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - John P. Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
155
|
Alfarouk KO, Alhoufie STS, Hifny A, Schwartz L, Alqahtani AS, Ahmed SBM, Alqahtani AM, Alqahtani SS, Muddathir AK, Ali H, Bashir AHH, Ibrahim ME, Greco MR, Cardone RA, Harguindey S, Reshkin SJ. Of mitochondrion and COVID-19. J Enzyme Inhib Med Chem 2021; 36:1258-1267. [PMID: 34107824 PMCID: PMC8205080 DOI: 10.1080/14756366.2021.1937144] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/20/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023] Open
Abstract
COVID-19, a pandemic disease caused by a viral infection, is associated with a high mortality rate. Most of the signs and symptoms, e.g. cytokine storm, electrolytes imbalances, thromboembolism, etc., are related to mitochondrial dysfunction. Therefore, targeting mitochondrion will represent a more rational treatment of COVID-19. The current work outlines how COVID-19's signs and symptoms are related to the mitochondrion. Proper understanding of the underlying causes might enhance the opportunity to treat COVID-19.
Collapse
Affiliation(s)
- Khalid Omer Alfarouk
- Research Center, Zamzam University College, Khartoum, Sudan
- Department of Evolutionary Pharmacology and Tumor Metabolism, Hala Alfarouk Cancer Center, Khartoum, Sudan
- Al-Ghad International College for Applied Medical Sciences, Al-Madinah Al-Munwarah, Saudi Arabia
| | - Sari T. S. Alhoufie
- Medical Laboratories Technology Department, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Munwarah, Saudi Arabia
| | | | | | - Ali S. Alqahtani
- College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | | | - Ali M. Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Saad S. Alqahtani
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | | | - Heyam Ali
- Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Adil H. H. Bashir
- Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | | | - Maria Raffaella Greco
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Rosa A. Cardone
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| | | | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
156
|
Jouzdani AF, Heidarimoghadam R, Hazhirkamal M, Ranjbar A. Nanoantioxidant/Antioxidant Therapy in 2019-nCoV: A New Approach to Reactive Oxygen Species Mechanisms. CURRENT DRUG THERAPY 2021. [DOI: 10.2174/1574885516666210719092931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The COVID-19 pandemic has caused serious concerns for people around the world. The
COVID-19 is associated with respiratory failure, generation of reactive oxygen species (ROS), and the
lack of antioxidants among patients. Specified ROS levels have an essential role as an adjuster of immunological
responses and virus cleaners, but excessive ROS will oxidize membrane lipids and cellular
proteins and quickly destroy virus-infected cells. It can also adversely damage normal cells in the
lungs and even the heart, resulting in multiple organ failures. Given the above, a highly potent antioxidant
therapy can be offered to reduce cardiac loss due to COVID-19. In modern medicine, nanoparticles
containing antioxidants can be used as a high-performance therapy in reducing oxidative stress in
the prevention and treatment of infectious diseases. This can provide a free and interactive tool to determine
whether antioxidants and nanoantioxidants can be administered for COVID-19. More research
and studies are needed to investigate and make definitive opinions about their medicinal uses.
Collapse
Affiliation(s)
- Ali Fathi Jouzdani
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Maryam Hazhirkamal
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Akram Ranjbar
- Nutrition Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
157
|
Li Z, Li Y, Li L, Mo X, Li S, Xie M, Zhan Y, Lin Y, Li Z, Xie M, Chen Z, Zhu A, Ying R, Yu L, Zhao J, Li SC, Cai W, Ye F. Alteration of the respiratory microbiome in COVID-19 patients with different severities. J Genet Genomics 2021; 49:258-261. [PMID: 34798357 PMCID: PMC8595322 DOI: 10.1016/j.jgg.2021.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Zhengtu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China
| | - Yinhu Li
- Department of Computer Sciences, City University of Hong Kong, Hong Kong, 999077, China
| | - Linghua Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Xiaoneng Mo
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Shaoqiang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China
| | - Mingzhou Xie
- Beijing YuanShengKangTai (ProtoDNA) Genetech Co. Ltd., Beijing, 100190, China
| | - Yangqing Zhan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China
| | - Ye Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China
| | - Zhun Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China
| | - Min Xie
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Zhaoming Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China
| | - Ruosu Ying
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Le Yu
- Beijing YuanShengKangTai (ProtoDNA) Genetech Co. Ltd., Beijing, 100190, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China
| | - Shuai Cheng Li
- Department of Computer Sciences, City University of Hong Kong, Hong Kong, 999077, China.
| | - Weiping Cai
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China.
| | - Feng Ye
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou, 510120, China.
| |
Collapse
|
158
|
Wu Y, Wang XH, Li XH, Song LY, Yu SL, Fang ZC, Liu YQ, Yuan LY, Peng CY, Zhang SY, Cheng W, Ma HC, Wang LF, Tang JM, Wang YF, Ji FY. Common mtDNA variations at C5178a and A249d/T6392C/G10310A decrease the risk of severe COVID-19 in a Han Chinese population from Central China. Mil Med Res 2021; 8:57. [PMID: 34724985 PMCID: PMC8558762 DOI: 10.1186/s40779-021-00351-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Mitochondria have been shown to play vital roles during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and coronavirus disease 2019 (COVID-19) development. Currently, it is unclear whether mitochondrial DNA (mtDNA) variants, which define mtDNA haplogroups and determine oxidative phosphorylation performance and reactive oxygen species production, are associated with COVID-19 risk. METHODS A population-based case-control study was conducted to compare the distribution of mtDNA variations defining mtDNA haplogroups between healthy controls (n = 615) and COVID-19 patients (n = 536). COVID-19 patients were diagnosed based on molecular diagnostics of the viral genome by qPCR and chest X-ray or computed tomography scanning. The exclusion criteria for the healthy controls were any history of disease in the month preceding the study assessment. MtDNA variants defining mtDNA haplogroups were identified by PCR-RFLPs and HVS-I sequencing and determined based on mtDNA phylogenetic analysis using Mitomap Phylogeny. Student's t-test was used for continuous variables, and Pearson's chi-squared test or Fisher's exact test was used for categorical variables. To assess the independent effect of each mtDNA variant defining mtDNA haplogroups, multivariate logistic regression analyses were performed to calculate the odds ratios (ORs) and 95% confidence intervals (CIs) with adjustments for possible confounding factors of age, sex, smoking and diseases (including cardiopulmonary diseases, diabetes, obesity and hypertension) as determined through clinical and radiographic examinations. RESULTS Multivariate logistic regression analyses revealed that the most common investigated mtDNA variations (> 10% in the control population) at C5178a (in NADH dehydrogenase subunit 2 gene, ND2) and A249d (in the displacement loop region, D-loop)/T6392C (in cytochrome c oxidase I gene, CO1)/G10310A (in ND3) were associated with a reduced risk of severe COVID-19 (OR = 0.590, 95% CI 0.428-0.814, P = 0.001; and OR = 0.654, 95% CI 0.457-0.936, P = 0.020, respectively), while A4833G (ND2), A4715G (ND2), T3394C (ND1) and G5417A (ND2)/C16257a (D-loop)/C16261T (D-loop) were related to an increased risk of severe COVID-19 (OR = 2.336, 95% CI 1.179-4.608, P = 0.015; OR = 2.033, 95% CI 1.242-3.322, P = 0.005; OR = 3.040, 95% CI 1.522-6.061, P = 0.002; and OR = 2.890, 95% CI 1.199-6.993, P = 0.018, respectively). CONCLUSIONS This is the first study to explore the association of mtDNA variants with individual's risk of developing severe COVID-19. Based on the case-control study, we concluded that the common mtDNA variants at C5178a and A249d/T6392C/G10310A might contribute to an individual's resistance to developing severe COVID-19, whereas A4833G, A4715G, T3394C and G5417A/C16257a/C16261T might increase an individual's risk of developing severe COVID-19.
Collapse
Affiliation(s)
- Yi Wu
- Department of Medical Biology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Xian-Hui Wang
- Institute of Biomedical Research, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Xi-Hua Li
- Department of Medical Biology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Li-Yuan Song
- Department of Medical Biology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Shi-Long Yu
- Institute of Human Respiratory Disease, Xinqiao Hospital, The Army Medical University (Third Military Medical University), 400037, Chongqing, China
| | - Zhi-Cheng Fang
- Department of Emergency Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Yu-Quan Liu
- Department of Geriatric Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Le-Yong Yuan
- Department of Immunology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Chun-Yan Peng
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Shen-Yi Zhang
- Department of Medical Biology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Wang Cheng
- Department of Medical Biology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Hong-Chao Ma
- Department of Medical Biology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Li-Feng Wang
- Department of Medical Biology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Jun-Ming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| | - Yun-Fu Wang
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| | - Fu-Yun Ji
- Department of Medical Biology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China. .,Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| |
Collapse
|
159
|
Abstract
In 2019 first reports about a new human coronavirus emerged, which causes common cold symptoms as well as acute respiratory distress syndrome. The virus was identified as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and severe thrombotic events including deep vein thrombosis, pulmonary embolism, and microthrombi emerged as additional symptoms. Heart failure, myocardial infarction, myocarditis, and stroke have also been observed. As main mediator of thrombus formation, platelets became one of the key aspects in SARS-CoV-2 research. Platelets may also directly interact with SARS-CoV-2 and have been shown to carry the SARS-CoV-2 virus. Platelets can also facilitate the virus uptake by secretion of the subtilisin-like proprotein convertase furin. Cleavage of the SARS-CoV-2 spike protein by furin enhances binding capabilities and virus entry into various cell types. In COVID-19 patients, platelet count differs between mild and serious infections. Patients with mild symptoms have a slightly increased platelet count, whereas thrombocytopenia is a hallmark of severe COVID-19 infections. Low platelet count can be attributed to platelet apoptosis and the incorporation of platelets into microthrombi (peripheral consumption) and severe thrombotic events. The observed excessive formation of thrombi is due to hyperactivation of platelets caused by the infection. Various factors have been suggested in the activation of platelets in COVID-19, such as hypoxia, vessel damage, inflammatory factors, NETosis, SARS-CoV-2 interaction, autoimmune reactions, and autocrine activation. COVID-19 does alter chemokine and cytokine plasma concentrations. Platelet chemokine profiles are altered in COVID-19 and contribute to the described chemokine storms observed in severely ill COVID-19 patients.
Collapse
Affiliation(s)
- Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls Universtität Tübingen, Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls Universtität Tübingen, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls Universtität Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls Universtität Tübingen, Tübingen, Germany
| |
Collapse
|
160
|
Kim WJ, Yum MS, Kim MJ, Jang HN, Ko TS. Stroke-Like Episodes Associated with Coronavirus Disease 2019 in a Child with MELAS Syndrome. ANNALS OF CHILD NEUROLOGY 2021. [DOI: 10.26815/acn.2021.00395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
161
|
Piotrowicz K, Gąsowski J, Michel JP, Veronese N. Post-COVID-19 acute sarcopenia: physiopathology and management. Aging Clin Exp Res 2021; 33:2887-2898. [PMID: 34328636 PMCID: PMC8323089 DOI: 10.1007/s40520-021-01942-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
In this review, we discuss the pathophysiologic and management aspects of acute sarcopenia in relation to SARS-CoV-2 infection. COVID-19 is as a multi-organ infectious disease characterized by a severe inflammatory and highly catabolic status, influencing the deep changes in the body build, especially the amount, structure, and function of skeletal muscles which would amount to acutely developed sarcopenia. Acute sarcopenia may largely impact patients’ in-hospital prognosis as well as the vulnerability to the post-COVID-19 functional and physical deterioration. The individual outcome of the COVID-19 and the degree of muscle mass and functional loss may be influenced by multiple factors, including the patient’s general pre-infection medical and functional condition, especially in older adults. This paper gathers the information about how the SARS-CoV-2 hyper-inflammatory involvement exacerbates the immunosenescence process, enhances the endothelial damage, and due to mitochondrial dysfunction and autophagy, induces myofibrillar breakdown and muscle degradation. The aftermath of these acute and complex immunological SARS-CoV-2-related phenomena, augmented by anosmia, ageusia and altered microbiota may lead to decreased food intake and exacerbated catabolism. Moreover, the imposed physical inactivity, lock-down, quarantine or acute hospitalization with bedrest would intensify the acute sarcopenia process. All these deleterious mechanisms must be swiftly put to a check by a multidisciplinary approach including nutritional support, early physical as well cardio-pulmonary rehabilitation, and psychological support and cognitive training. The proposed holistic and early management of COVID-19 patients appears essential to minimize the disastrous functional outcomes of this disease and allow avoiding the long COVID-19 syndrome.
Collapse
Affiliation(s)
- Karolina Piotrowicz
- Department of Internal Medicine and Gerontology, Faculty of Medicine, Jagiellonian University Medical College, 2 Jakubowskiego St., building I, 5th floor, 30-688, Kraków, Poland
| | - Jerzy Gąsowski
- Department of Internal Medicine and Gerontology, Faculty of Medicine, Jagiellonian University Medical College, 2 Jakubowskiego St., building I, 5th floor, 30-688, Kraków, Poland.
| | | | - Nicola Veronese
- Department of Internal Medicine, Geriatrics Section, University of Palermo, Palermo, Italy
| |
Collapse
|
162
|
Huang T, Zhang T, Jiang X, Li A, Su Y, Bian Q, Wu H, Lin R, Li N, Cao H, Ling D, Wang J, Tabata Y, Gu Z, Gao J. Iron oxide nanoparticles augment the intercellular mitochondrial transfer-mediated therapy. SCIENCE ADVANCES 2021; 7:eabj0534. [PMID: 34586849 PMCID: PMC8480934 DOI: 10.1126/sciadv.abj0534] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/30/2021] [Indexed: 05/24/2023]
Abstract
The transfer of mitochondria between cells has recently been revealed as a spontaneous way to protect the injured cells. However, the utilization of this natural transfer process for disease treatment is so far limited by its unsatisfactory transfer efficiency and selectivity. Here, we demonstrate that iron oxide nanoparticles (IONPs) can augment the intercellular mitochondrial transfer from human mesenchymal stem cells (hMSCs) selectively to diseased cells, owing to the enhanced formation of connexin 43–containing gap junctional channels triggered by ionized IONPs. In a mouse model of pulmonary fibrosis, the IONP-engineered hMSCs achieve a remarkable mitigation of fibrotic progression because of the promoted intercellular mitochondrial transfer, with no serious safety issues identified. The present study reports a potential method of using IONPs to enable hMSCs for efficient and safe transfer of mitochondria to diseased cells to restore mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Ting Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinchi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ai Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanqin Su
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiong Bian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghui Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ruyi Lin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ni Li
- Department of Cardiothoracic Surgery, Ningbo Medical Center, Lihuili Hospital Affiliated to Ningbo University, Ningbo, Zhejiang 315041, China
| | - Hongcui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Daishun Ling
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinqiang Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Westlake Laboratory of Life Sciences and Biomedicine, Zhejiang, China
| |
Collapse
|
163
|
Lemos AEG, Silva GR, Gimba ERP, Matos ADR. Susceptibility of lung cancer patients to COVID-19: A review of the pandemic data from multiple nationalities. Thorac Cancer 2021; 12:2637-2647. [PMID: 34435733 PMCID: PMC8520793 DOI: 10.1111/1759-7714.14067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 01/08/2023] Open
Abstract
Several studies have highlighted that cancer patients tend to be more susceptible to develop severe infection and to die from COVID-19. Certain medical conditions such as immunosuppression, presence of comorbidities, and underlying pulmonary damage are possible determinants of disease severity, especially in lung cancer patients. While recent studies have shown that lung cancer is one of the most prevalent tumor types among COVID-19 cancer patients, we still have an incomplete view of how data from several countries work as a whole. The aim of this review was to investigate COVID-19 prevalence in lung cancer patient cohorts and their probability to develop severe illness and death when compared to nonlung cancer patients from multiple nationalities, including countries that have been the epicenters of the pandemic. We also focus on some intrinsic lung cancer features that might influence COVID-19 outcomes. An integrative view of the susceptibility of lung cancer patients might be especially relevant to assist physicians in evaluating the risks of COVID-19 in these patients, and to foster better decisions on treatment delay.
Collapse
Affiliation(s)
- Ana Emília Goulart Lemos
- Department of Physiology and PharmacologyBiomedical Institute, Federal Fluminense University (UFF)NiteroiBrazil
- National School of Public Health Sergio Arouca, Department of Epidemiology and Quantitative Methods in HealthOswaldo Cruz Foundation (FIOCRUZ)Rio de JaneiroBrazil
- Cellular and Molecular Oncobiology Program, Research CentreNational Cancer Institute (INCA)Rio de JaneiroBrazil
| | - Gabriela Ribeiro Silva
- Department of Physiology and PharmacologyBiomedical Institute, Federal Fluminense University (UFF)NiteroiBrazil
- Cellular and Molecular Oncobiology Program, Research CentreNational Cancer Institute (INCA)Rio de JaneiroBrazil
| | - Etel Rodrigues Pereira Gimba
- Department of Physiology and PharmacologyBiomedical Institute, Federal Fluminense University (UFF)NiteroiBrazil
- Cellular and Molecular Oncobiology Program, Research CentreNational Cancer Institute (INCA)Rio de JaneiroBrazil
- Institute of Humanities and Health, Department of Natural SciencesFederal Fluminense University (UFF)Rio das OstrasBrazil
| | - Aline da Rocha Matos
- Oswaldo Cruz Institute, Respiratory and Measles Viruses Laboratory/SARS‐CoV‐2 Reference, Laboratory, MoHWorld Health Organization (WHO), FIOCRUZRio de JaneiroBrazil
| |
Collapse
|
164
|
Microbiota and Its Impact on the Immune System in COVID-19-A Narrative Review. J Clin Med 2021; 10:jcm10194537. [PMID: 34640553 PMCID: PMC8509181 DOI: 10.3390/jcm10194537] [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: 09/03/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 01/08/2023] Open
Abstract
The microbiota is of interest for the development of a therapeutic strategy against SARS-CoV-2 coronavirus disease 2019 (COVID-19) due to its impact on the host immune system. Proven communications of the gut microbiota with the pulmonary microbiota (gut-lung axis) and the pathway of neural connections between the gut and brain (gut-brain axis) may be important in the face of the pandemic. SARS-CoV-2 was shown to affect almost all organs because of the presence of a host receptor known as angiotensin converting enzyme 2 (ACE2). The ACE2 receptor is mainly present in the brush border of intestinal enterocytes, ciliary cells, and type II alveolar epithelial cells in the lungs. The transport function of ACE2 has been linked to the ecology of gut microbes in the digestive tract, suggesting that COVID-19 may be related to the gut microbiota. The severity of COVID-19 may be associated with a number of comorbidities, such as hypertension, diabetes, obesity, and/or old age; therefore, attention is also paid to multiple morbidities and the modulation of microbiota through comorbidities and medications. This paper reviews the research in the context of the state of the intestinal microbiota and its impact on the cells of the immune system during the SARS-CoV-2 pandemic.
Collapse
|
165
|
van der Walt G, Lindeque JZ, Mason S, Louw R. Sub-Cellular Metabolomics Contributes Mitochondria-Specific Metabolic Insights to a Mouse Model of Leigh Syndrome. Metabolites 2021; 11:metabo11100658. [PMID: 34677373 PMCID: PMC8537744 DOI: 10.3390/metabo11100658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Direct injury of mitochondrial respiratory chain (RC) complex I by Ndufs4 subunit mutations results in complex I deficiency (CID) and a progressive encephalomyopathy, known as Leigh syndrome. While mitochondrial, cytosolic and multi-organelle pathways are known to be involved in the neuromuscular LS pathogenesis, compartment-specific metabolomics has, to date, not been applied to murine models of CID. We thus hypothesized that sub-cellular metabolomics would be able to contribute organelle-specific insights to known Ndufs4 metabolic perturbations. To that end, whole brains and skeletal muscle from late-stage Ndufs4 mice and age/sex-matched controls were harvested for mitochondrial and cytosolic isolation. Untargeted 1H-NMR and semi-targeted LC-MS/MS metabolomics was applied to the resulting cell fractions, whereafter important variables (VIPs) were selected by univariate statistics. A predominant increase in multiple targeted amino acids was observed in whole-brain samples, with a more prominent effect at the mitochondrial level. Similar pathways were implicated in the muscle tissue, showing a greater depletion of core metabolites with a compartment-specific distribution, however. The altered metabolites expectedly implicate altered redox homeostasis, alternate RC fueling, one-carbon metabolism, urea cycling and dysregulated proteostasis to different degrees in the analyzed tissues. A first application of EDTA-chelated magnesium and calcium measurement by NMR also revealed tissue- and compartment-specific alterations, implicating stress response-related calcium redistribution between neural cell compartments, as well as whole-cell muscle magnesium depletion. Altogether, these results confirm the ability of compartment-specific metabolomics to capture known alterations related to Ndufs4 KO and CID while proving its worth in elucidating metabolic compartmentalization in said pathways that went undetected in the diluted whole-cell samples previously studied.
Collapse
|
166
|
Jabczyk M, Nowak J, Hudzik B, Zubelewicz-Szkodzińska B. Diet, Probiotics and Their Impact on the Gut Microbiota during the COVID-19 Pandemic. Nutrients 2021; 13:3172. [PMID: 34579048 PMCID: PMC8465654 DOI: 10.3390/nu13093172] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 01/09/2023] Open
Abstract
SARS-CoV-2 infection is associated with diverse clinical manifestations, immune dysfunction, and gut microbiota alterations. The nutritional and biochemical quality of one's diet can influence the intestinal microbiota, which may play a role in the defense mechanisms against potential pathogens, by promoting a wide variety of immune-host interactions. In the COVID-19 pandemic, besides the development of pharmacological therapies, a healthy balanced diet, rich with food-derived antioxidants, may be a useful strategy. Many studies demonstrated that vitamins and probiotic therapies have positive effects on the treatment and prevention of oxidative stress and inflammation in COVID-19. The ecology of the gut microbiota in the digestive tract has been linked to the transport function of the host receptor known as angiotensin converting enzyme 2 (ACE2), suggesting that COVID-19 may be related to the gut microbiota. The angiotensin converting enzyme (ACE), and its receptor (ACE2), play central roles in modulating the renin-angiotensin system (RAS). In addition, ACE2 has functions that act independently of the RAS. ACE2 is the receptor for the SARS coronavirus, and ACE2 is essential for the expression of neutral amino acid transporters in the gut. In this context, ACE2 modulates innate immunity and influences the composition of the gut microbiota. Malnutrition is one of the leading underlying causes of morbidity and mortality worldwide and, including comorbidities, may be a major cause of worse outcomes and higher mortality among COVID-19 patients. This paper reviews the research on dietary components, with particular emphasis on vitamins, antioxidants, and probiotic therapies, and their impacts on the intestinal microbiota's diversity during the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Marzena Jabczyk
- Department of Nutrition-Related Disease Prevention, Faculty of Health Sciences in Bytom, Medical University of Silesia, Piekarska 18 Street, 41-902 Bytom, Poland; (M.J.); (B.Z.-S.)
| | - Justyna Nowak
- Department of Cardiovascular Disease Prevention, Faculty of Health Sciences in Bytom, Medical University of Silesia, Piekarska 18 Street, 41-902 Bytom, Poland;
| | - Bartosz Hudzik
- Department of Cardiovascular Disease Prevention, Faculty of Health Sciences in Bytom, Medical University of Silesia, Piekarska 18 Street, 41-902 Bytom, Poland;
- Silesian Center for Heart Diseases, Third Department of Cardiology, Faculty of Medical Science in Zabrze, Medical University of Silesia, 41-800 Zabrze, Poland
| | - Barbara Zubelewicz-Szkodzińska
- Department of Nutrition-Related Disease Prevention, Faculty of Health Sciences in Bytom, Medical University of Silesia, Piekarska 18 Street, 41-902 Bytom, Poland; (M.J.); (B.Z.-S.)
| |
Collapse
|
167
|
Santos AF, Póvoa P, Paixão P, Mendonça A, Taborda-Barata L. Changes in Glycolytic Pathway in SARS-COV 2 Infection and Their Importance in Understanding the Severity of COVID-19. Front Chem 2021; 9:685196. [PMID: 34568275 PMCID: PMC8461303 DOI: 10.3389/fchem.2021.685196] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
COVID-19 is an infectious disease caused by Coronavirus 2 (SARS-CoV-2) that may lead to a severe acute respiratory syndrome. Such syndrome is thought to be related, at least in part, to a dysregulation of the immune system which involves three main components: hyperactivity of the innate immune system; decreased production of type 1 Interferons (IFN) by SARS-CoV-2-infected cells, namely respiratory epithelial cells and macrophages; and decreased numbers of both CD4+ and particularly CD8+ T cells. Herein, we describe how excessive activation of the innate immune system and the need for viral replication in several cells of the infected organism promote significant alterations in cells' energy metabolism (glucose metabolism), which may underlie the poor prognosis of the disease in severe situations. When activated, cells of the innate immune system reprogram their metabolism, and increase glucose uptake to ensure secretion of pro-inflammatory cytokines. Changes in glucose metabolism are also observed in pulmonary epithelial cells, contributing to dysregulation of cytokine synthesis and inflammation of the pulmonary epithelium. Controlling hyperglycolysis in critically ill patients may help to reduce the exaggerated production of pro-inflammatory cytokines and optimise the actions of the adaptive immune system. In this review, we suggest that the administration of non-toxic concentrations of 2-deoxy-D-glucose, the use of GLUT 1 inhibitors, of antioxidants such as vitamin C in high doses, as well as the administration of N-acetylcysteine in high doses, may be useful complementary therapeutic strategies for these patients, as suggested by some clinical trials and/ or reports. Overall, understanding changes in the glycolytic pathway associated with COVID-19 infection can help to find new forms of treatment for this disease.
Collapse
Affiliation(s)
- Adalberto Fernandes Santos
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
- CICS-UBI Health Sciences Research Centre, Universidade da Beira Interior, Lisbon, Portugal
- Department of Teaching and Research of Biochemistry, Faculty of Medicine, Agostinho Neto University, Luanda, Angola
| | - Pedro Póvoa
- Polyvalent Intensive Care Unit, Centro Hospitalar de Lisboa Ocidental, Hospital de Sao Francisco Xavier, Lisbon, Portugal
- Comprehensive Health Research Center–CHRC, NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
- Center for Clinical Epidemiology and Research Unit of Clinical Epidemiology, OUH Odense University Hospital, Odense, Denmark
| | - Paulo Paixão
- Comprehensive Health Research Center–CHRC, NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
- Laboratório de Patologia Clínica–SYNLAB, Hospital da Luz, Lisbon, Portugal
| | - António Mendonça
- CICS-UBI Health Sciences Research Centre, Universidade da Beira Interior, Lisbon, Portugal
- Department of Chemistry, Faculty of Sciences, University of Beira Interior, Covilhã, Portugal
| | - Luís Taborda-Barata
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
- CICS-UBI Health Sciences Research Centre, Universidade da Beira Interior, Lisbon, Portugal
- Department of Immunoallergology, Cova da Beira University Hospital Centre, Covilhã, Portugal
| |
Collapse
|
168
|
Valdés-Aguayo JJ, Garza-Veloz I, Badillo-Almaráz JI, Bernal-Silva S, Martínez-Vázquez MC, Juárez-Alcalá V, Vargas-Rodríguez JR, Gaeta-Velasco ML, González-Fuentes C, Ávila-Carrasco L, Martinez-Fierro ML. Mitochondria and Mitochondrial DNA: Key Elements in the Pathogenesis and Exacerbation of the Inflammatory State Caused by COVID-19. ACTA ACUST UNITED AC 2021; 57:medicina57090928. [PMID: 34577851 PMCID: PMC8471487 DOI: 10.3390/medicina57090928] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/21/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
Background and Objectives. The importance of mitochondria in inflammatory pathologies, besides providing energy, is associated with the release of mitochondrial damage products, such as mitochondrial DNA (mt-DNA), which may perpetuate inflammation. In this review, we aimed to show the importance of mitochondria, as organelles that produce energy and intervene in multiple pathologies, focusing mainly in COVID-19 and using multiple molecular mechanisms that allow for the replication and maintenance of the viral genome, leading to the exacerbation and spread of the inflammatory response. The evidence suggests that mitochondria are implicated in the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which forms double-membrane vesicles and evades detection by the cell defense system. These mitochondrion-hijacking vesicles damage the integrity of the mitochondrion’s membrane, releasing mt-DNA into circulation and triggering the activation of innate immunity, which may contribute to an exacerbation of the pro-inflammatory state. Conclusions. While mitochondrial dysfunction in COVID-19 continues to be studied, the use of mt-DNA as an indicator of prognosis and severity is a potential area yet to be explored.
Collapse
Affiliation(s)
- José J. Valdés-Aguayo
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (J.J.V.-A.); (I.G.-V.); (J.I.B.-A.); (M.C.M.-V.); (V.J.-A.); (J.R.V.-R.); (L.Á.-C.)
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (J.J.V.-A.); (I.G.-V.); (J.I.B.-A.); (M.C.M.-V.); (V.J.-A.); (J.R.V.-R.); (L.Á.-C.)
| | - José I. Badillo-Almaráz
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (J.J.V.-A.); (I.G.-V.); (J.I.B.-A.); (M.C.M.-V.); (V.J.-A.); (J.R.V.-R.); (L.Á.-C.)
| | - Sofia Bernal-Silva
- Microbiology Department, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Avenida Venustiano Carranza 2405, San Luis Potosí 78210, Mexico;
| | - Maria C. Martínez-Vázquez
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (J.J.V.-A.); (I.G.-V.); (J.I.B.-A.); (M.C.M.-V.); (V.J.-A.); (J.R.V.-R.); (L.Á.-C.)
| | - Vladimir Juárez-Alcalá
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (J.J.V.-A.); (I.G.-V.); (J.I.B.-A.); (M.C.M.-V.); (V.J.-A.); (J.R.V.-R.); (L.Á.-C.)
| | - José R. Vargas-Rodríguez
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (J.J.V.-A.); (I.G.-V.); (J.I.B.-A.); (M.C.M.-V.); (V.J.-A.); (J.R.V.-R.); (L.Á.-C.)
| | - María L. Gaeta-Velasco
- Hospital General de Zacatecas “Luz González Cosío”, Circuito Ciudad Gobierno 410, Col. Ciudad Gobierno, Zacatecas 98160, Mexico; (M.L.G.-V.); (C.G.-F.)
| | - Carolina González-Fuentes
- Hospital General de Zacatecas “Luz González Cosío”, Circuito Ciudad Gobierno 410, Col. Ciudad Gobierno, Zacatecas 98160, Mexico; (M.L.G.-V.); (C.G.-F.)
| | - Lorena Ávila-Carrasco
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (J.J.V.-A.); (I.G.-V.); (J.I.B.-A.); (M.C.M.-V.); (V.J.-A.); (J.R.V.-R.); (L.Á.-C.)
| | - Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S., Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (J.J.V.-A.); (I.G.-V.); (J.I.B.-A.); (M.C.M.-V.); (V.J.-A.); (J.R.V.-R.); (L.Á.-C.)
- Correspondence: ; Tel.: +52-(492)-925669 (ext. 4511)
| |
Collapse
|
169
|
John A, Ali K, Marsh H, Reddy PH. Can healthy lifestyle reduce disease progression of Alzheimer's during a global pandemic of COVID-19? Ageing Res Rev 2021; 70:101406. [PMID: 34242809 PMCID: PMC8259043 DOI: 10.1016/j.arr.2021.101406] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/05/2021] [Accepted: 07/05/2021] [Indexed: 12/13/2022]
Abstract
The novel coronavirus disease 2019 (COVID-19) has pushed the medical system to its breaking point. While the virus does not discriminate, the elderly and those with comorbidities, including hypertension severe obesity, diabetes mellitus, coronary disease, pneumonia and dementia, are at a greater risk for adverse outcomes due to COVID-19. While many people navigate their new normal, the question of what the long-lasting effects of the pandemic may be, lingers. To investigate how vulnerable populations are affected by the pandemic, we focused on Alzheimer's disease, a vector to understanding how the virus has impacted AD progression and risk via aging. By assessing the effect of COVID-19 on AD patients, we explore genetics, metabolism, and lifestyle factors in both COVID-19 and Alzheimer's disease that can work synergistically to precipitate adverse outcomes. This article also discusses how age-related conditions and/or age-related comorbidities susceptible to COVID-19. We also discuss possible healthy lifestyle factors reduce and/or combat COVID-19 now and in the future.
Collapse
Affiliation(s)
- Albin John
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Kiran Ali
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Harrison Marsh
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
170
|
Altay O, Arif M, Li X, Yang H, Aydın M, Alkurt G, Kim W, Akyol D, Zhang C, Dinler‐Doganay G, Turkez H, Shoaie S, Nielsen J, Borén J, Olmuscelik O, Doganay L, Uhlén M, Mardinoglu A. Combined Metabolic Activators Accelerates Recovery in Mild-to-Moderate COVID-19. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101222. [PMID: 34180141 PMCID: PMC8420376 DOI: 10.1002/advs.202101222] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/19/2021] [Indexed: 05/02/2023]
Abstract
COVID-19 is associated with mitochondrial dysfunction and metabolic abnormalities, including the deficiencies in nicotinamide adenine dinucleotide (NAD+ ) and glutathione metabolism. Here it is investigated if administration of a mixture of combined metabolic activators (CMAs) consisting of glutathione and NAD+ precursors can restore metabolic function and thus aid the recovery of COVID-19 patients. CMAs include l-serine, N-acetyl-l-cysteine, nicotinamide riboside, and l-carnitine tartrate, salt form of l-carnitine. Placebo-controlled, open-label phase 2 study and double-blinded phase 3 clinical trials are conducted to investigate the time of symptom-free recovery on ambulatory patients using CMAs. The results of both studies show that the time to complete recovery is significantly shorter in the CMA group (6.6 vs 9.3 d) in phase 2 and (5.7 vs 9.2 d) in phase 3 trials compared to placebo group. A comprehensive analysis of the plasma metabolome and proteome reveals major metabolic changes. Plasma levels of proteins and metabolites associated with inflammation and antioxidant metabolism are significantly improved in patients treated with CMAs as compared to placebo. The results show that treating patients infected with COVID-19 with CMAs lead to a more rapid symptom-free recovery, suggesting a role for such a therapeutic regime in the treatment of infections leading to respiratory problems.
Collapse
Affiliation(s)
- Ozlem Altay
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
- Department of Clinical MicrobiologyDr Sami Ulus Training and Research HospitalUniversity of Health SciencesAnkara06080Turkey
| | - Muhammad Arif
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
| | - Xiangyu Li
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
| | - Hong Yang
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
| | - Mehtap Aydın
- Department of Infectious DiseasesUmraniye Training and Research HospitalUniversity of Health SciencesIstanbul34766Turkey
| | - Gizem Alkurt
- Genomic Laboratory (GLAB)Umraniye Training and Research HospitalUniversity of Health SciencesIstanbul34766Turkey
| | - Woonghee Kim
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
| | - Dogukan Akyol
- Genomic Laboratory (GLAB)Umraniye Training and Research HospitalUniversity of Health SciencesIstanbul34766Turkey
| | - Cheng Zhang
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation TechnologiesMinistry of EducationZhengzhou UniversityZhengzhouHenan450001P. R. China
| | - Gizem Dinler‐Doganay
- Department of Molecular Biology and GeneticsIstanbul Technical UniversityIstanbul34469Turkey
| | - Hasan Turkez
- Department of Medical BiologyFaculty of MedicineAtatürk UniversityErzurum25240Turkey
| | - Saeed Shoaie
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonSE1 1ULUK
| | - Jens Nielsen
- Department of Biology and Biological EngineeringChalmers University of TechnologyGothenburgSE‐41296Sweden
| | - Jan Borén
- Department of Molecular and Clinical MedicineUniversity of Gothenburg and Sahlgrenska University Hospital GothenburgGothenburgSE‐41345Sweden
| | - Oktay Olmuscelik
- Department of Internal MedicineIstanbul Medipol UniversityBagcılarIstanbul34214Turkey
| | - Levent Doganay
- Department of GastroenterologyUmraniye Training and Research HospitalUniversity of Health SciencesIstanbul34766Turkey
| | - Mathias Uhlén
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
| | - Adil Mardinoglu
- Science for Life LaboratoryKTH—Royal Institute of TechnologyStockholmSE‐100 44Sweden
- Centre for Host‐Microbiome InteractionsFaculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonSE1 1ULUK
| |
Collapse
|
171
|
Paul BD, Lemle MD, Komaroff AL, Snyder SH. Redox imbalance links COVID-19 and myalgic encephalomyelitis/chronic fatigue syndrome. Proc Natl Acad Sci U S A 2021; 118:e2024358118. [PMID: 34400495 PMCID: PMC8403932 DOI: 10.1073/pnas.2024358118] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Although most patients recover from acute COVID-19, some experience postacute sequelae of severe acute respiratory syndrome coronavirus 2 infection (PASC). One subgroup of PASC is a syndrome called "long COVID-19," reminiscent of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). ME/CFS is a debilitating condition, often triggered by viral and bacterial infections, leading to years-long debilitating symptoms including profound fatigue, postexertional malaise, unrefreshing sleep, cognitive deficits, and orthostatic intolerance. Some are skeptical that either ME/CFS or long COVID-19 involves underlying biological abnormalities. However, in this review, we summarize the evidence that people with acute COVID-19 and with ME/CFS have biological abnormalities including redox imbalance, systemic inflammation and neuroinflammation, an impaired ability to generate adenosine triphosphate, and a general hypometabolic state. These phenomena have not yet been well studied in people with long COVID-19, and each of them has been reported in other diseases as well, particularly neurological diseases. We also examine the bidirectional relationship between redox imbalance, inflammation, energy metabolic deficits, and a hypometabolic state. We speculate as to what may be causing these abnormalities. Thus, understanding the molecular underpinnings of both PASC and ME/CFS may lead to the development of novel therapeutics.
Collapse
Affiliation(s)
- Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205;
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | - Anthony L Komaroff
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02120
| | - Solomon H Snyder
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205;
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
172
|
Keane C, Coalter M, Martin-Loeches I. Immune System Disequilibrium-Neutrophils, Their Extracellular Traps, and COVID-19-Induced Sepsis. Front Med (Lausanne) 2021; 8:711397. [PMID: 34485339 PMCID: PMC8416266 DOI: 10.3389/fmed.2021.711397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Equilibrium within the immune system can often determine the fate of its host. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) pandemic. Immune dysregulation remains one of the main pathophysiological components of SARS-CoV-2-associated organ injury, with over-activation of the innate immune system, and induced apoptosis of adaptive immune cells. Here, we provide an overview of the innate immune system, both in general and relating to COVID-19. We specifically discuss "NETosis," the process of neutrophil release of their extracellular traps, which may be a more recently described form of cell death that is different from apoptosis, and how this may propagate organ dysfunction in COVID-19. We complete this review by discussing Stem Cell Therapies in COVID-19 and emerging COVID-19 phenotypes, which may allow for more targeted therapy in the future. Finally, we consider the array of potential therapeutic targets in COVID-19, and associated therapeutics.
Collapse
Affiliation(s)
- Colm Keane
- Department of Anaesthesia and Intensive Care, St. James's Hospital, Dublin, Ireland
- Multidisciplinary Intensive Care Research Organization (MICRO), Trinity College Dublin, Dublin, Ireland
| | - Matthew Coalter
- Department of Anaesthesia and Intensive Care, St. James's Hospital, Dublin, Ireland
| | - Ignacio Martin-Loeches
- Department of Anaesthesia and Intensive Care, St. James's Hospital, Dublin, Ireland
- Multidisciplinary Intensive Care Research Organization (MICRO), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
173
|
Pandey RK, Pandey RK, Shukla SS, Pandey P. A review on corona virus and treatment approaches with Allium sativam. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021; 7:159. [PMID: 34395639 PMCID: PMC8353433 DOI: 10.1186/s43094-021-00310-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/04/2021] [Indexed: 11/13/2022] Open
Abstract
Background Recently reported cases of Covid-19 globally remind us that new diseases are coming while we are unable to provide the treatment for the same. The entire world is facing this viral attack; deaths are increasing day by day as well as infected patients too. Today, in the period of this disease, can we go to the shelter of our
traditional medicines? Main body In this article, we have taken medicines related to corona and conceptualized their mechanism, which gave us a chance to understand Garlic's mechanism of action, how Garlic can be a weapon in the lane with this disease. This article also tells how we can treat new diseases with our traditional herbs if no modern medicine has been discovered yet. Conclusion The present review is based on the structure of the virus and the targeted site for the drug discovery process with important constituents of Allium sativam. The review work also explains the allicin chemical constituent of Allium sativam which has targeted therapeutic sites related to Covid-19.
Collapse
Affiliation(s)
- Rupesh Kumar Pandey
- Department of Pharmacology KSCP, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh India
| | | | | | - Priyanka Pandey
- Swami Vivekanand College of Pharmacy, Indore, Madhya Pradesh India
| |
Collapse
|
174
|
Ardatskaya MD, Butorova LI, Kalashnikova MA, Nugaeva NR, Ovchinnikov YV, Oynotkinova OS, Pavlov AI, Plavnik RG, Sayutina EV, Topchiy TВ, Trunova SN. [Gastroenterological symptoms in COVID-19 patients with mild severity of the disease: opportunities to optimize antidiarrheal therapy]. TERAPEVT ARKH 2021; 93:923-931. [PMID: 36286887 DOI: 10.26442/00403660.2021.08.201020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The novel coronavirus infection COVID-19 can be manifested by damage to the organs of the gastrointestinal tract (GIT). Damage to the gastrointestinal tract by the SARS-CoV-2 virus leads to a violation of the microbial-tissue complex of the mucous membrane of the digestive tract. A common gastroenterological manifestation of COVID-19 is diarrhea. AIM Study of the clinical features of gastroenterological disorders and the possibility of optimizing the treatment of diarrheal syndrome in patients with COVID-19 with a mild form of viral infection. MATERIALS AND METHODS The observation group consisted of 230 patients with mild COVID-19: K-group (n=115) with respiratory symptoms, I group (n=115) with gastrointestinal manifestations in combination and without signs of respiratory damage. In order to compare the effectiveness of treatment of diarrheal syndrome, patients of group I are randomized into 2 subgroups: Ia (n=58) prebiotic treatment (Zacofalk) and Ib (n=57) enterosorbents. RESULTS The development of gastrointestinal symptoms with SARS-CoV-2 infection is significantly more often noted in comorbid patients (67%). Gastrointestinal symptoms were dominated by diarrhea (93.9%) and flatulence (76.5%), in 1/3 of patients they were the first manifestos of infection. It was established that in 98.4% of patients of group I (against 42.6% of the K-group) signs of infectious intoxication were detected. In patients with gastrointestinal lesions, an elongation of the febrile period by 91.5 days was noted, a later (6 days) verification of the viral etiology of the disease. It was found that in patients of group I, the regression of clinical symptoms, the duration of viral disease, the dynamics of antibody formation, the prognosis for the development of IBS-like disorders in the post-infectious period depended on the treatment. In patients taking (Zacofalk), these indicators were significantly better. CONCLUSION In mild cases, to reduce the severity of viral intestinal damage, for effective relief of intestinal symptoms, to reduce the risk of IBS-like symptoms, it is advisable to prescribe (Zacofalk) in an initial dose of 3 tablets per day.
Collapse
Affiliation(s)
| | - L I Butorova
- Central State Medical Academy of the President of the Russian Federation
| | - M A Kalashnikova
- Central Clinical Hospital for Rehabilitation Treatment of the President of the Russian Federation
| | | | | | - O S Oynotkinova
- Lomonosov Moscow State University
- Research Institute for Healthcare Organization and Medical Management
| | - A I Pavlov
- Vishnevsky 3rd Central Military Clinical Hospital
| | | | - E V Sayutina
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - T В Topchiy
- Central State Medical Academy of the President of the Russian Federation
| | | |
Collapse
|
175
|
Lower Serum Angiotensin-Converting Enzyme Level in Relation to Hyperinflammation and Impaired Antiviral Immune Response Contributes to Progression of COVID-19 Infection. Infect Dis Ther 2021; 10:2431-2446. [PMID: 34387835 PMCID: PMC8361819 DOI: 10.1007/s40121-021-00513-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/26/2021] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION As a homologue of the angiotensin-converting enzyme (ACE), angiotensin-converting enzyme 2 (ACE2) has been identified as the main receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) invasion. We aimed to investigate the role of serum ACE in predicting the coronavirus disease 2019 (COVID-19) disease progression and the underlying mechanisms. METHODS We retrospectively enrolled 120 patients with confirmed COVID-19 who underwent serum ACE detection on admission. The clinical characteristics and laboratory findings during hospitalization were evaluated dynamically to identify the potential risk factors for disease progression. RESULTS ACE level was demonstrated as one of the independent risk factors. Patients with ACE level ≤ 33.5 U/L showed a higher cumulative virus RNA detection rate, elevated pro-inflammatory mediators levels, declined lymphocyte count, and decreased SARS-CoV-2-specific antibodies than those with ACE level > 33.5 U/L. CONCLUSION Lower serum ACE levels in relation to delayed virus elimination, hyperinflammatory condition, and impaired host antiviral immune responses contribute to disease progression of COVID-19.
Collapse
|
176
|
Mitochondrial DNA in innate immune responses against infectious diseases. Biochem Soc Trans 2021; 48:2823-2838. [PMID: 33155647 DOI: 10.1042/bst20200687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/18/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022]
Abstract
Mitochondrial DNA (mtDNA) can initiate an innate immune response when mislocalized in a compartment other than the mitochondrial matrix. mtDNA plays significant roles in regulating mitochondrial dynamics as well as mitochondrial unfolded protein response (UPR). The mislocalized extra-mtDNA can elicit innate immune response via cGAS-STING (cyclic GMP-AMP synthase-stimulator of interferon genes) pathway, inducing the expression of the interferon-stimulated genes (ISGs). Also, cytosolic damaged mtDNA is cleared up by various pathways which are responsible for participating in the activation of inflammatory responses. Four pathways of extra-mitochondrial mtDNA clearance are highlighted in this review - the inflammasome activation mechanism, neutrophil extracellular traps formation, recognition by Toll-like receptor 9 and transfer of mtDNA between cells packaged into extracellular vesicles. Anomalies in these pathways are associated with various diseases. We posit our review in the present pandemic situation and discuss how mtDNA elicits innate immune responses against different viruses and bacteria. This review gives a comprehensive picture of the role of extra-mitochondrial mtDNA in infectious diseases and speculates that research towards its understanding would help establish its therapeutic potential.
Collapse
|
177
|
Jesuthasan A, Massey F, Manji H, Zandi MS, Wiethoff S. Emerging potential mechanisms and predispositions to the neurological manifestations of COVID-19. J Neurol Sci 2021; 428:117608. [PMID: 34391037 PMCID: PMC8332920 DOI: 10.1016/j.jns.2021.117608] [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: 05/20/2021] [Revised: 07/10/2021] [Accepted: 08/01/2021] [Indexed: 12/23/2022]
Abstract
A spectrum of neurological disease associated with COVID-19 is becoming increasingly apparent. However, the mechanisms behind these manifestations remain poorly understood, significantly hindering their management. The present review subsequently attempts to address the evolving molecular, cellular and systemic mechanisms of NeuroCOVID, which we have classified as the acute and long-term neurological effects of COVID-19. We place particular emphasis on cerebrovascular, demyelinating and encephalitic presentations, which have been reported. Several mechanisms are presented, especially the involvement of a "cytokine storm". We explore the genetic and demographic factors that may predispose individuals to NeuroCOVID. The increasingly evident long-term neurological effects are also presented, including the impact of the virus on cognition, autonomic function and mental wellbeing, which represent an impending burden on already stretched healthcare services. We subsequently reinforce the need for cautious surveillance, especially for those with predisposing factors, with effective clinical phenotyping, appropriate investigation and, if possible, prompt treatment. This will be imperative to prevent downstream neurological sequelae, including those related to the long COVID phenotypes that are being increasingly recognised.
Collapse
Affiliation(s)
- Aaron Jesuthasan
- University College Hospital, University College London Hospitals NHS Foundation Trust, London, UK.
| | - Flavia Massey
- University College London Medical School, Gower Street, London, UK
| | - Hadi Manji
- National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Michael S Zandi
- National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Sarah Wiethoff
- UCL Institute of Neurology, Queen Square, London, UK; Klinik für Neurologie mit Institut für Translationale Neurologie, Albert Schweitzer Campus 1, Gebäude A1, D-48149 Münster, Germany
| |
Collapse
|
178
|
Valenti D, Vacca RA, Moro L, Atlante A. Mitochondria Can Cross Cell Boundaries: An Overview of the Biological Relevance, Pathophysiological Implications and Therapeutic Perspectives of Intercellular Mitochondrial Transfer. Int J Mol Sci 2021; 22:8312. [PMID: 34361078 PMCID: PMC8347886 DOI: 10.3390/ijms22158312] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 01/07/2023] Open
Abstract
Mitochondria are complex intracellular organelles traditionally identified as the powerhouses of eukaryotic cells due to their central role in bioenergetic metabolism. In recent decades, the growing interest in mitochondria research has revealed that these multifunctional organelles are more than just the cell powerhouses, playing many other key roles as signaling platforms that regulate cell metabolism, proliferation, death and immunological response. As key regulators, mitochondria, when dysfunctional, are involved in the pathogenesis of a wide range of metabolic, neurodegenerative, immune and neoplastic disorders. Far more recently, mitochondria attracted renewed attention from the scientific community for their ability of intercellular translocation that can involve whole mitochondria, mitochondrial genome or other mitochondrial components. The intercellular transport of mitochondria, defined as horizontal mitochondrial transfer, can occur in mammalian cells both in vitro and in vivo, and in physiological and pathological conditions. Mitochondrial transfer can provide an exogenous mitochondrial source, replenishing dysfunctional mitochondria, thereby improving mitochondrial faults or, as in in the case of tumor cells, changing their functional skills and response to chemotherapy. In this review, we will provide an overview of the state of the art of the up-to-date knowledge on intercellular trafficking of mitochondria by discussing its biological relevance, mode and mechanisms underlying the process and its involvement in different pathophysiological contexts, highlighting its therapeutic potential for diseases with mitochondrial dysfunction primarily involved in their pathogenesis.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM)-CNR, Via G. Amendola122/O, 70126 Bari, Italy; (R.A.V.); (L.M.)
| | | | | | - Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM)-CNR, Via G. Amendola122/O, 70126 Bari, Italy; (R.A.V.); (L.M.)
| |
Collapse
|
179
|
Chikhale R, Sinha SK, Wanjari M, Gurav NS, Ayyanar M, Prasad S, Khanal P, Dey YN, Patil RB, Gurav SS. Computational assessment of saikosaponins as adjuvant treatment for COVID-19: molecular docking, dynamics, and network pharmacology analysis. Mol Divers 2021; 25:1889-1904. [PMID: 33492566 PMCID: PMC7829483 DOI: 10.1007/s11030-021-10183-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/06/2021] [Indexed: 12/29/2022]
Abstract
Saikosaponins are major biologically active triterpenoids, usually as glucosides, isolated from Traditional Chinese Medicines (TCM) such as Bupleurum spp., Heteromorpha spp., and Scrophularia scorodonia with their antiviral and immunomodulatory potential. This investigation presents molecular docking, molecular dynamics simulation, and free energy calculation studies of saikosaponins as adjuvant therapy in the treatment for COVID19. Molecular docking studies for 23 saikosaponins on the crystal structures of the extracellular domains of human lnterleukin-6 receptor (IL6), human Janus Kinase-3 (JAK3), and dehydrogenase domain of Cylindrospermum stagnale NADPH-oxidase 5 (NOX5) were performed, and selected protein-ligand complexes were subjected to 100 ns molecular dynamics simulations. The molecular dynamics trajectories were subjected to free energy calculation by the MM-GBSA method. Molecular docking and molecular dynamics simulation studies revealed that IL6 in complex with Saikosaponin_U and Saikosaponin_V, JAK3 in complex with Saikosaponin_B4 and Saikosaponin_I, and NOX5 in complex with Saikosaponin_BK1 and Saikosaponin_C have good docking and molecular dynamics profiles. However, the Janus Kinase-3 is the best interacting partner for the saikosaponin compounds. The network pharmacology analysis suggests saikosaponins interact with the proteins CAT Gene CAT (Catalase) and Checkpoint kinase 1 (CHEK1); both of these enzymes play a major role in cell homeostasis and DNA damage during infection, suggesting a possible improvement in immune response toward COVID-19.
Collapse
Affiliation(s)
- Rupesh Chikhale
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Saurabh K Sinha
- Department of Pharmaceutical Sciences, Mohanlal Shukhadia University, Udaipur, Rajasthan, 313 001, India
| | - Manish Wanjari
- Regional Ayurveda Research Institute for Drug Development, Gwalior, Madhya Pradesh, 474009, India
| | - Nilambari S Gurav
- PES's Rajaram and Tarabai Bandekar College of Pharmacy, Goa University, Ponda, Goa, 403401, India
| | - Muniappan Ayyanar
- Department of Botany, A. Veeriya Vandayar Memorial Sri Pushpam College (Autonomous), Affiliated To Bharathidasan University, Poondi, Thanjavur, 613 503, India
| | - Satyendra Prasad
- Department of Pharmaceutical Sciences, R.T.M. University, Nagpur, Maharashtra, 440033, India
| | - Pukar Khanal
- Department of Pharmacology and Toxicology, KLE College of Pharmacy Belagavi, KLE Academy of Higher Education and Research (KAHER), Belagavi, 590010, India
| | - Yadu Nandan Dey
- School of Pharmaceutical Technology, Adamas University, Kolkata, West Bengal, 700126, India
| | - Rajesh B Patil
- Sinhgad Technical Education Society's, Smt. Kashibai Navale College of Pharmacy, Pune, Maharashtra, India.
| | - Shailendra S Gurav
- Department of Pharmacognosy and Phytochemistry, Goa College of Pharmacy, Goa University, Panaji, Goa, 403 001, India.
| |
Collapse
|
180
|
Mate A, Reyes-Goya C, Santana-Garrido Á, Sobrevia L, Vázquez CM. Impact of maternal nutrition in viral infections during pregnancy. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166231. [PMID: 34343638 PMCID: PMC8325560 DOI: 10.1016/j.bbadis.2021.166231] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/05/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022]
Abstract
Other than being a physiological process, pregnancy is a condition characterized by major adaptations of maternal endocrine and metabolic homeostasis that are necessary to accommodate the fetoplacental unit. Unfortunately, all these systemic, cellular, and molecular changes in maternal physiology also make the mother and the fetus more prone to adverse outcomes, including numerous alterations arising from viral infections. Common infections during pregnancy that have long been recognized as congenitally and perinatally transmissible to newborns include toxoplasmosis, rubella, cytomegalovirus, and herpes simplex viruses (originally coined as ToRCH infections). In addition, enterovirus, parvovirus B19, hepatitis virus, varicella-zoster virus, human immunodeficiency virus, Zika and Dengue virus, and, more recently, coronavirus infections including Middle Eastern respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) infections (especially the novel SARS-CoV-2 responsible for the ongoing COVID-19 pandemic), constitute relevant targets for current research on maternal-fetal interactions in viral infections during pregnancy. Appropriate maternal education from preconception to the early postnatal period is crucial to promote healthy pregnancies in general and to prevent and/or reduce the impact of viral infections in particular. Specifically, an adequate lifestyle based on proper nutrition plans and feeding interventions, whenever possible, might be crucial to reduce the risk of virus-related gestational diseases and accompanying complications in later life. Here we aim to provide an overview of the emerging literature addressing the impact of nutrition in the context of potentially harmful viral infections during pregnancy.
Collapse
Affiliation(s)
- Alfonso Mate
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Epidemiología Clínica y Riesgo Cardiovascular, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain.
| | - Claudia Reyes-Goya
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | - Álvaro Santana-Garrido
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Epidemiología Clínica y Riesgo Cardiovascular, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Luis Sobrevia
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Medical School (Faculty of Medicine), São Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (UMCG), 9713GZ Groningen, the Netherlands
| | - Carmen M Vázquez
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; Epidemiología Clínica y Riesgo Cardiovascular, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| |
Collapse
|
181
|
da Silva KN, Gobatto ALN, Costa-Ferro ZSM, Cavalcante BRR, Caria ACI, de Aragão França LS, Nonaka CKV, de Macêdo Lima F, Lopes-Pacheco M, Rocco PRM, de Freitas Souza BS. Is there a place for mesenchymal stromal cell-based therapies in the therapeutic armamentarium against COVID-19? Stem Cell Res Ther 2021; 12:425. [PMID: 34315546 PMCID: PMC8314259 DOI: 10.1186/s13287-021-02502-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023] Open
Abstract
The COVID-19 pandemic, caused by the rapid global spread of the novel coronavirus (SARS-CoV-2), has caused healthcare systems to collapse and led to hundreds of thousands of deaths. The clinical spectrum of COVID-19 is not only limited to local pneumonia but also represents multiple organ involvement, with potential for systemic complications. One year after the pandemic, pathophysiological knowledge has evolved, and many therapeutic advances have occurred, but mortality rates are still elevated in severe/critical COVID-19 cases. Mesenchymal stromal cells (MSCs) can exert immunomodulatory, antiviral, and pro-regenerative paracrine/endocrine actions and are therefore promising candidates for MSC-based therapies. In this review, we discuss the rationale for MSC-based therapies based on currently available preclinical and clinical evidence of safety, potential efficacy, and mechanisms of action. Finally, we present a critical analysis of the risks, limitations, challenges, and opportunities that place MSC-based products as a therapeutic strategy that may complement the current arsenal against COVID-19 and reduce the pandemic's unmet medical needs.
Collapse
Affiliation(s)
- Kátia Nunes da Silva
- Goncalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
| | | | - Zaquer Suzana Munhoz Costa-Ferro
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
| | - Bruno Raphael Ribeiro Cavalcante
- Goncalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia, 40296-710, Brazil
| | - Alex Cleber Improta Caria
- Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Luciana Souza de Aragão França
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
| | - Carolina Kymie Vasques Nonaka
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
| | | | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macêdo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science and Technology, and Innovation, Rio de Janeiro, Brazil
| | - Bruno Solano de Freitas Souza
- Goncalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rua Waldemar Falcão, 121, Candeal, Salvador, Bahia, 40296-710, Brazil.
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil.
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil.
| |
Collapse
|
182
|
Fišar Z, Ľupták M, Hroudová J. Little in vitro effect of remdesivir on mitochondrial respiration and monoamine oxidase activity in isolated mitochondria. Toxicol Lett 2021; 350:143-151. [PMID: 34311047 DOI: 10.1016/j.toxlet.2021.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/13/2022]
Abstract
Remdesivir (RDV) is a novel antiviral drug whose mitochondrial effects are not well known. In vitro effects of RDV on the mitochondrial respiration, individual respiratory complexes, and the activity of monoamine oxidase (MAO-A and MAO-B) were measured in isolated mitochondria. At micromolar RDV concentrations, minimal or no inhibitory effects on the studied mitochondrial enzymes were found. At very high concentrations of RDV, there was partial inhibition of complex I- (IC50 675 μmol/L, residual activity 39.4 %) and complex II-linked (IC50 81.8 μmol/L, residual activity 40.7 %) respiration, without inhibition of complex IV-linked respiration, and partial inhibition both of MAO-A (IC50 26.6 μmol/L, residual activity 35.2 %) and MAO-B (IC50 89.8 μmol/L, residual activity 34.0 %) activity. Individual respiratory complexes (I, II + III, and IV) were partially inhibited at a high drug concentration. The active metabolite of RDV (GS-443902) had very little effect on mitochondrial oxygen consumption rate with residual activity of 87.0 % for complex I-linked respiration, 90.3 % for complex II-linked respiration, and with no inhibition of complex IV-linked respiration. In conclusion, measurement of the effect of RDV and its active metabolite on isolated mitochondria shows that there is very little direct effect on mitochondrial respiration occurs at therapeutic drug concentration.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Ke Karlovu 11, 121 28, Prague 2, Czech Republic.
| | - Matej Ľupták
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Institute of Pharmacology, Albertov 4, 128 00, Prague 2, Czech Republic
| | - Jana Hroudová
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Ke Karlovu 11, 121 28, Prague 2, Czech Republic
| |
Collapse
|
183
|
Clavario P, De Marzo V, Lotti R, Barbara C, Porcile A, Russo C, Beccaria F, Bonavia M, Bottaro LC, Caltabellotta M, Chioni F, Santangelo M, Hautala AJ, Griffo R, Parati G, Corrà U, Porto I. Cardiopulmonary exercise testing in COVID-19 patients at 3 months follow-up. Int J Cardiol 2021; 340:113-118. [PMID: 34311011 PMCID: PMC8302817 DOI: 10.1016/j.ijcard.2021.07.033] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/29/2021] [Accepted: 07/14/2021] [Indexed: 12/16/2022]
Abstract
Background Long-term effects of Coronavirus Disease of 2019 (COVID-19) are of utmost relevance. We aimed to determine: 1) the functional capacity of COVID-19 survivors by cardiopulmonary exercise testing (CPET); 2) the characteristics associated with cardiopulmonary exercise testing (CPET) performance; 3) the safety and tolerability of CPET. Methods We prospectively enrolled consecutive patients with laboratory-confirmed COVID-19 from Azienda Sanitaria Locale 3, Genoa. Three months after hospital discharge a complete clinical evaluation, trans-thoracic echocardiography, CPET, pulmonary function tests, and dominant leg extension (DLE) maximal strength measurement were performed. Results From the 225 patients discharged alive from March to November 2020, we excluded 12 incomplete/missing cases and 13 unable to perform CPET, leading to a final cohort of 200. Median percent-predicted peak oxygen uptake (%pVO2) was 88% (78.3–103.1). Ninety-nine (49.5%) patients had %pVO2 below, whereas 101 (50.5%) above the 85% predicted value. Among the 99 patients with reduced %pVO2, 61 (61%) had a normal anaerobic threshold: of these, 9(14.8%) had respiratory, 21(34.4%) cardiac, and 31(50.8%) non-cardiopulmonary reasons for exercise limitation. Inerestingly, 80% of patients experienced at least one disabling symtpom, not related to %pVO2 or functional capacity. Multivariate linear regression showed percent-predicted forced expiratory volume in one-second(β = 5.29,p = 0.023), percent-predicted diffusing capacity of lungs for carbon monoxide(β = 6.31,p = 0.001), and DLE maximal strength(β = 14.09,p = 0.008) to be independently associated with pVO2. No adverse event was reported during or after CPET, and no involved health professional developed COVID-19. Conclusions At three months after discharge, about 1/3rd of COVID-19 survivors show functional limitations, mainly explained by muscular impairment, calling for future research to identify patients at higher risk of long-term effects that may benefit from careful surveillance and targeted rehabilitation.
Collapse
Affiliation(s)
- Piero Clavario
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Vincenzo De Marzo
- Cardiology Unit, DICATOV - Cardiothoracic and Vascular Department, IRCCS San Martino Hospital, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Roberta Lotti
- Cardiology Unit, DICATOV - Cardiothoracic and Vascular Department, IRCCS San Martino Hospital, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Cristina Barbara
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Annalisa Porcile
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Carmelo Russo
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Federica Beccaria
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Marco Bonavia
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Luigi Carlo Bottaro
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Marta Caltabellotta
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Flavia Chioni
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Monica Santangelo
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Arto J Hautala
- Faculty of Sports and Health Sciences, University of Jyväskylä, Finland
| | - Raffaele Griffo
- Cardiac Rehabilitation Center of Genoa, Azienda Sanitaria Locale, ASL 3 Genovese, Genoa, Italy
| | - Gianfranco Parati
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Ugo Corrà
- IRCCS, Istituti Clinici Scientifici Maugeri, Care and Research Institute, Department of Cardiac Rehabilitation, Veruno-Novara, Italy
| | - Italo Porto
- Cardiology Unit, DICATOV - Cardiothoracic and Vascular Department, IRCCS San Martino Hospital, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy.
| |
Collapse
|
184
|
Pasini E, Corsetti G, Romano C, Scarabelli TM, Chen-Scarabelli C, Saravolatz L, Dioguardi FS. Serum Metabolic Profile in Patients With Long-Covid (PASC) Syndrome: Clinical Implications. Front Med (Lausanne) 2021; 8:714426. [PMID: 34368201 PMCID: PMC8339407 DOI: 10.3389/fmed.2021.714426] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/29/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Many patients who have been suffering by Covid-19 suffer of long-Covid syndrome, with symptoms of fatigue and muscular weakness that characterize post-acute sequelae SARS-CoV-2 infection (PASC). However, there is limited knowledge about the molecular pathophysiology, and about the serum profile of these patients. Methods: We studied the blood serum profile of 75 selected patients, with previous confirmed Covid-19, 2 months after hospital discharge, who reported new-onset fatigue, muscle weakness and/or dyspnea not present prior to the virus infection and independently from concomitant diseases and/or clinical conditions. Results: All patients had very high serum concentrations of ferritin and D-Dimer. 87 and 72% of patients had clinically significant low levels of hemoglobin and albumin, respectively. Seventy three percentage had elevations in erythrocyte sedimentation rate and CRP. Twenty seven percentage had elevations in LDH. Conclusions: The co-existence of patient symptoms along with blood markers of coagulation, protein disarrangement and inflammation suggests ongoing alterations in the metabolism, promoting an inflammatory/hypercatabolic state which maintains a vicious circles implicated in the persistence of PASC. The persistence of altered D-Dimer levels raises the possibility of long-term risks of thromboembolic disease. All these markers levels should be accurately evaluated in the long-term follow-up, with individualized consideration for prophylactic nutritional, anti-inflammatory and/or anticoagulant therapy if indicated.
Collapse
Affiliation(s)
- Evasio Pasini
- Division of Cardiac Rehabilitation, Scientific Clinical Institutes Maugeri Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Lumezzane, Italy
| | - Giovanni Corsetti
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Claudia Romano
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Tiziano M Scarabelli
- Center for Heart and Vessel Preclinical Studies, St. John Hospital and Medical Center, Wayne State University, Detroit, MI, United States
| | - Carol Chen-Scarabelli
- Division of Cardiology, Richmond Veterans Affairs Medical Center, Richmond, VA, United States
| | - Louis Saravolatz
- Department of Medicine at St. John Hospital, Wayne State University, Detroit, MI, United States
| | | |
Collapse
|
185
|
Investigating the potential mechanisms of depression induced-by COVID-19 infection in patients. J Clin Neurosci 2021; 91:283-287. [PMID: 34373041 PMCID: PMC8289699 DOI: 10.1016/j.jocn.2021.07.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/01/2021] [Accepted: 07/16/2021] [Indexed: 12/19/2022]
Abstract
The new coronavirus (COVID-19) has emerged now in the world as a pandemic. The SARS-CoV-2 infection causes variant common symptoms, such as dry cough, tiredness, dyspnea, fever, myalgia, chills, headache, chest pain, and conjunctivitis. Different organs may be affected by COVID-19, such as the respiratory system, gastrointestinal tract, kidneys, and CNS. However, the information about the COVID-19 infection in the CNS is insufficient. We do know that the virus can enter the central nervous system (CNS) via different routes, causing symptoms such as dizziness, headache, seizures, loss of consciousness, and depression. Depression is the most common disorder among all neurological symptoms following COVID-19 infection, although the mechanism of COVID-19-induced depression is not yet clear. The aim of the present study is to investigate the probable mechanisms of COVID-19-induced depression. The reasons for depression in infected patients may be due to social and pathological factors including social quarantine, economic problems, stress, changes in the HPA axis, inflammation due to the entry of proinflammatory cytokines into the CNS, production of inflammatory cytokines by microglia, mitochondrial disorders, damage to the hippocampus, and malnutrition. By evaluating different factors involved in COVID-19-induced depression, we have concluded that depression can be minimized by controlling stress, preventing the cytokine storm with appropriate anti-inflammatory drugs, and proper nutrition.
Collapse
|
186
|
Yousof SM, Tanvir I, Kolieb E, Atta R. Work Stress, Dysbiosis, and Immune Dysregulation: The Interconnected Triad in COVID-19 Infection in the Medical Team Staff - A Mini-Review. J Microsc Ultrastruct 2021; 10:147-153. [PMID: 36687328 PMCID: PMC9846923 DOI: 10.4103/jmau.jmau_9_21] [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: 01/27/2021] [Accepted: 04/17/2021] [Indexed: 01/25/2023] Open
Abstract
The COVID-19 pandemic has hit most of the communities around the globe. Earlier researches have reported the psychological effects of pandemics either on the general populations or on specific communities such as students and health professionals. A scanty number of papers have focused on the interaction among complex factors underlying the pathogenesis of the disease. In this review, we aimed to integrate the accessible data about the possible mechanistic processes predisposing to COVID-19 infection in the health professions. We summarized these factors as "stress, microbiota, and immunity triad." We utilized the PubMed database, Google, and Google Scholar search engines to search the literature related to combinations of these keywords: "pandemics, COVID-19, coronavirus, SARS-CoV2;" "gut microbiota, gut-lung axis, dysbiosis, nutrition;" "work stress, workload, health workers, health professions, and medical team;" and "immunity, cytokine storm, and viral load." We detected no discussions combining the suggested triad concerning the medical team personnel. We cast light, for the first time to our knowledge, on the potential pathogenic role of "stress, microbiota, and immunity triad" in COVID-19-infected health workers.
Collapse
Affiliation(s)
- Shimaa Mohammad Yousof
- Department of Medical Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt,Department of Medical Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt,Address for correspondence: Dr. Shimaa Mohammad Yousof, Department of Medical Physiology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia.
Department of Medical Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt. E-mail:
| | - Imrana Tanvir
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Rabigh Branch, Rabigh, Saudi Arabia
| | - Eman Kolieb
- Department of Medical Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Rasha Atta
- Department of Medical Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
187
|
Cai M, Du B, Si Y, Miao J, Ge J, Zhang J, Song J, Bao H. Knockdown of VDAC1 alleviates the cognitive dysfunction secondary to sepsis-associated encephalopathy. Am J Transl Res 2021; 13:7538-7555. [PMID: 34377234 PMCID: PMC8340252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/13/2021] [Indexed: 06/13/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is a serious and diffuse cerebral dysregulation with a high morbidity and mortality caused by sepsis. Mitophagy plays an important role in SAE, and microglial phagocytosis of apoptotic cells (efferocytosis) is the core of the brain regenerative response. Voltage dependent anion channel (VDAC1) is an important regulator of mitophagy. However, it remains unknown whether VDAC1 influences SAE progression by regulating mitophagy and efferocytosis. Herein, we explored the mechanism where knockdown of VDAC1 alleviated the cognitive dysfunction caused by sepsis-associated encephalopathy and further elucidated the underlying molecular mechanisms. SAE model in mice was established through caecal ligation and puncture (CLP). The increased mitophagy and decreased efferocytosis were observed by the transmission electron microscope (TEM) in the SAE model. Besides, immunoblot tests showed an interaction between autophagy and efferocytosis. Further behavior tests and TEM results indicated that knockdown of VDAC1 alleviated the cognitive dysfunction by decreasing the autophagy and increasing the efferocytosis in a PINK1/Parkin-dependent manner. Based on these results, we conclude that knockdown of VDAC1 alleviates the cognitive dysfunction in the CLP-induced SAE mouse model.
Collapse
Affiliation(s)
- Mengmeng Cai
- Department of Anesthesiology, Affiliated Nanjing Hospital of Nanjing Medical University (Nanjing First Hospital)Nanjing, Jiangsu Province, China
| | - Boxiang Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Yanna Si
- Department of Anesthesiology, Affiliated Nanjing Hospital of Nanjing Medical University (Nanjing First Hospital)Nanjing, Jiangsu Province, China
| | - Juanjuan Miao
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Jianyun Ge
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Jiejie Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Jie Song
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Hongguang Bao
- Department of Anesthesiology, Affiliated Nanjing Hospital of Nanjing Medical University (Nanjing First Hospital)Nanjing, Jiangsu Province, China
| |
Collapse
|
188
|
Burtscher J, Burtscher M, Millet GP. The central role of mitochondrial fitness on antiviral defenses: An advocacy for physical activity during the COVID-19 pandemic. Redox Biol 2021; 43:101976. [PMID: 33932869 PMCID: PMC8062414 DOI: 10.1016/j.redox.2021.101976] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are central regulators of cellular metabolism, most known for their role in energy production. They can be "enhanced" by physical activity (including exercise), which increases their integrity, efficiency and dynamic adaptation to stressors, in short "mitochondrial fitness". Mitochondrial fitness is closely associated with cardiorespiratory fitness and physical activity. Given the importance of mitochondria in immune functions, it is thus not surprising that cardiorespiratory fitness is also an integral determinant of the antiviral host defense and vulnerability to infection. Here, we first briefly review the role of physical activity in viral infections. We then summarize mitochondrial functions that are relevant for the antiviral immune response with a particular focus on the current Coronavirus Disease (COVID-19) pandemic and on innate immune function. Finally, the modulation of mitochondrial and cardiorespiratory fitness by physical activity, aging and the chronic diseases that represent the most common comorbidities of COVID-19 is discussed. We conclude that a high mitochondrial - and related cardiorespiratory - fitness should be considered as protective factors for viral infections, including COVID-19. This assumption is corroborated by reduced mitochondrial fitness in many established risk factors of COVID-19, like age, various chronic diseases or obesity. We argue for regular analysis of the cardiorespiratory fitness of COVID-19 patients and the promotion of physical activity - with all its associated health benefits - as preventive measures against viral infection.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland.
| | | | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland
| |
Collapse
|
189
|
Notz Q, Herrmann J, Schlesinger T, Helmer P, Sudowe S, Sun Q, Hackler J, Roeder D, Lotz C, Meybohm P, Kranke P, Schomburg L, Stoppe C. Clinical Significance of Micronutrient Supplementation in Critically Ill COVID-19 Patients with Severe ARDS. Nutrients 2021; 13:nu13062113. [PMID: 34203015 PMCID: PMC8235175 DOI: 10.3390/nu13062113] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/02/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
The interplay between inflammation and oxidative stress is a vicious circle, potentially resulting in organ damage. Essential micronutrients such as selenium (Se) and zinc (Zn) support anti-oxidative defense systems and are commonly depleted in severe disease. This single-center retrospective study investigated micronutrient levels under Se and Zn supplementation in critically ill patients with COVID-19 induced acute respiratory distress syndrome (ARDS) and explored potential relationships with immunological and clinical parameters. According to intensive care unit (ICU) standard operating procedures, patients received 1.0 mg of intravenous Se daily on top of artificial nutrition, which contained various amounts of Se and Zn. Micronutrients, inflammatory cytokines, lymphocyte subsets and clinical data were extracted from the patient data management system on admission and after 10 to 14 days of treatment. Forty-six patients were screened for eligibility and 22 patients were included in the study. Twenty-one patients (95%) suffered from severe ARDS and 14 patients (64%) survived to ICU discharge. On admission, the majority of patients had low Se status biomarkers and Zn levels, along with elevated inflammatory parameters. Se supplementation significantly elevated Se (p = 0.027) and selenoprotein P levels (SELENOP; p = 0.016) to normal range. Accordingly, glutathione peroxidase 3 (GPx3) activity increased over time (p = 0.021). Se biomarkers, most notably SELENOP, were inversely correlated with CRP (rs = −0.495), PCT (rs = −0.413), IL-6 (rs = −0.429), IL-1β (rs = −0.440) and IL-10 (rs = −0.461). Positive associations were found for CD8+ T cells (rs = 0.636), NK cells (rs = 0.772), total IgG (rs = 0.493) and PaO2/FiO2 ratios (rs = 0.504). In addition, survivors tended to have higher Se levels after 10 to 14 days compared to non-survivors (p = 0.075). Sufficient Se and Zn levels may potentially be of clinical significance for an adequate immune response in critically ill patients with severe COVID-19 ARDS.
Collapse
Affiliation(s)
- Quirin Notz
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
- Correspondence: ; Tel.: +49-(0)931-201-30001
| | - Johannes Herrmann
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
| | - Tobias Schlesinger
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
| | - Philipp Helmer
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
| | | | - Qian Sun
- Institute of Experimental Endocrinology, Charité-Universitätsmedizin Berlin, D-10115 Berlin, Germany; (Q.S.); (J.H.); (L.S.)
| | - Julian Hackler
- Institute of Experimental Endocrinology, Charité-Universitätsmedizin Berlin, D-10115 Berlin, Germany; (Q.S.); (J.H.); (L.S.)
| | - Daniel Roeder
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
| | - Christopher Lotz
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
| | - Patrick Meybohm
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
| | - Peter Kranke
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
| | - Lutz Schomburg
- Institute of Experimental Endocrinology, Charité-Universitätsmedizin Berlin, D-10115 Berlin, Germany; (Q.S.); (J.H.); (L.S.)
| | - Christian Stoppe
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, D-97080 Wuerzburg, Germany; (J.H.); (T.S.); (P.H.); (D.R.); (C.L.); (P.M.); (P.K.); (C.S.)
| |
Collapse
|
190
|
Cárdenas-Rodríguez N, Bandala C, Vanoye-Carlo A, Ignacio-Mejía I, Gómez-Manzo S, Hernández-Cruz EY, Pedraza-Chaverri J, Carmona-Aparicio L, Hernández-Ochoa B. Use of Antioxidants for the Neuro-Therapeutic Management of COVID-19. Antioxidants (Basel) 2021; 10:971. [PMID: 34204362 PMCID: PMC8235474 DOI: 10.3390/antiox10060971] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus Disease 2019 (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is an emergent infectious disease that has caused millions of deaths throughout the world. COVID-19 infection's main symptoms are fever, cough, fatigue, and neurological manifestations such as headache, myalgias, anosmia, ageusia, impaired consciousness, seizures, and even neuromuscular junctions' disorders. In addition, it is known that this disease causes a series of systemic complications such as adverse respiratory distress syndrome, cardiac injury, acute kidney injury, and liver dysfunction. Due to the neurological symptoms associated with COVID-19, damage in the central nervous system has been suggested as well as the neuroinvasive potential of SARS-CoV-2. It is known that CoV infections are associated with an inflammation process related to the imbalance of the antioxidant system; cellular changes caused by oxidative stress contribute to brain tissue damage. Although anti-COVID-19 vaccines are under development, there is no specific treatment for COVID-19 and its clinical manifestations and complications; only supportive treatments with immunomodulators, anti-vascular endothelial growth factors, modulating drugs, statins, or nutritional supplements have been used. In the present work, we analyzed the potential of antioxidants as adjuvants for the treatment of COVID-19 and specifically their possible role in preventing or decreasing the neurological manifestations and neurological complications present in the disease.
Collapse
Affiliation(s)
- Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secreatría de Salud, Ciudad de México 04530, Mexico; (A.V.-C.); (L.C.-A.)
| | - Cindy Bandala
- Division de Neurociencias, Instituto Nacional de Rehabilitación, Secretaría de Salud, Ciudad de México 14389, Mexico;
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - América Vanoye-Carlo
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secreatría de Salud, Ciudad de México 04530, Mexico; (A.V.-C.); (L.C.-A.)
| | - Iván Ignacio-Mejía
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, SEDENA, Ciudad de México 11200, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | | | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, UNAM, Ciudad de México 04150, Mexico; (E.Y.H.-C.); (J.P.-C.)
| | - Liliana Carmona-Aparicio
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secreatría de Salud, Ciudad de México 04530, Mexico; (A.V.-C.); (L.C.-A.)
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México 06720, Mexico;
| |
Collapse
|
191
|
Katz-Agranov N, Zandman-Goddard G. Autoimmunity and COVID-19 - The microbiotal connection. Autoimmun Rev 2021; 20:102865. [PMID: 34118455 PMCID: PMC8189735 DOI: 10.1016/j.autrev.2021.102865] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023]
Abstract
Background and aims The novel SARS-CoV-2 has been rattling the world since its outbreak in December 2019, leading to the COVID-19 pandemic. The learning curve of this new virus has been steep, with a global scientific community desperate to learn how the virus is transmitted, how it replicates, why it causes such a wide spectrum of disease manifestations, resulting in none or few symptoms in some. Others are burdened by an intense immune response that resembles the cytokine storm syndrome (CSS), which leads to severe disease manifestations, often complicated by fatal acute respiratory distress syndrome and death. Research efforts have been focusing on finding effective cures and vaccinations for this virus. The presence of SARS-CoV-2 in the gastrointestinal (GI) tract, represented by several GI manifestations, has led to its investigation as a target for the virus and as an indicator of disease severity. The response of the microbiome (which is heavily linked to immunity) to the novel SARS-CoV-2 virus, and its role in igniting the exaggerated immune response has therefore become a focus of interest. The objective of our study was to gather the data connecting between the microbiome, the GI tract and COVID-19 and to investigate whether these reported alterations in the gut microbiome bear any resemblance to those seen in lupus, the prototypical autoimmune disease. Confirming such changes may become the steppingstone to potential therapies that may prevent transmission, progression and immune related manifestations of COVID-19, via manipulation of the gut microbiota. Methods We performed an extensive literature review, utilizing the Pubmed search engine and Google Scholar for studies evaluating the microbiome in COVID-19 patients and compared results with studies evaluating the microbiome in lupus. We searched for the terms: microbiome, dysbiosis, COVID-19, SARS-CoV-2, gastrointestinal as well as lupus and autoimmune. While there were hundreds of articles which referred to gastrointestinal manifestations in COVID-19, to date only 4 studies investigated the gastrointestinal microbiome in this setting. We compared the similarities between microbiome of COVID-19 patients and lupus patients. Results We found that there are several similar processes of immune dysregulation in patients with COVID-19 and in those with lupus, with several other alterations seen in other pathological states. Some of these similarities include loss of microbiota biodiversity, increased representation of pathobionts, which are microbes associated with inflammation and disease (i.e Proteobacteria) and a relative decrease of symbionts, which are protective microbes, associated with anti-inflammatory properties (i.e Lactobacillus). Compromise to the intestinal barrier has also been reported in both. Conclusions We conclude that the gastrointestinal tract contributes to the disease manifestations in COVID-19. Whether gastrointestinal dysbiosis is the cause or effect of gastrointestinal manifestations and several severe systemic manifestations, which may be the response to an increased pro-inflammatory environment, is still debatable and warrants further investigation. Given the resemblance of the microbiome in COVID-19 patients to that seen in lupus patients, it becomes clearer why several therapies used in autoimmune conditions are currently under investigation for the treatment of COVID-19 patients. Moreover, these findings should promote further investigating the utility of manipulation of the microbiome, via nutritional supplementation or even fecal transplantations, interventions that may alter the course of the disease, and potentially prevent disease transmission at low cost and low risk.
Collapse
Affiliation(s)
- Nurit Katz-Agranov
- Department of Medicine, Saint Elizabeth's Medical Center, Boston, MA, USA; Tufts University School of Medicine, Boston, MA, USA.
| | - Gisele Zandman-Goddard
- Department of Medicine C, Wolfson Medical Center, Holon, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
192
|
Karkhanei B, Talebi Ghane E, Mehri F. Evaluation of oxidative stress level: total antioxidant capacity, total oxidant status and glutathione activity in patients with COVID-19. New Microbes New Infect 2021; 42:100897. [PMID: 34026228 PMCID: PMC8127525 DOI: 10.1016/j.nmni.2021.100897] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), as a dangerous global pandemic, has led to high morbidity and mortality in all countries. There is a lot of evidence for the possible role of oxidative stress in COVID-19. In the present study, we aimed to measure the levels of glutathione (GSH), total antioxidant capacity (TAC) and total oxidant status (TOS) in the serum of patients with COVID-19. A total of 96 individuals with and without COVID-19 were enrolled and divided into four groups, including hospitalised group in non–intensive care units (non-ICU) (n = 35), hospitalised group in intensive care units with endotracheal intubation (EI) (ICU with EI) (n = 19), hospitalised group in intensive care units without endotracheal intubation (ICU without EI) (n = 24) and healthy people without COVID-19 disease as our control group (n = 18). The present study revealed that the TOS level was significantly lower in the group of control (p = 0.001), and level of GSH remarkably increased in the patients' groups (p < 0.001). TAC activity in non-ICU group of patients had no significant difference in comparison with the control group. However, in hospitalised patients' groups in the ICU with and without EI this activity was significantly different from the control group (p < 0.001). Moreover, there was a significant relationship between the levels of TOS, GSH and TAC with blood oxygen saturation (SpO2), fever, duration of hospitalisation and the prognosis of this disease (p < 0.001). Area under the curve (CI, 95%) of TOS, TAC and GSH-C to predict death among patients were, respectively, 0.907 (0.841, 0.973), 0.735 (0.626, 0.843) and 0.820 (0.725, 0.914). Receiver operating characteristic curve analysis showed that TOS, TAC and GSH-C have the potential specificity and sensitivity to distinguish between alive and dead patients. We found that elevated levels of oxidative stress and reduction of antioxidant indices can aggravate disease's severity in hospitalised patients with COVID-19. Therefore, it can be suggested to apply antioxidant agents as one of the effective therapeutic strategies in these groups.
Collapse
Affiliation(s)
- B Karkhanei
- Department of Anesthesiology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - E Talebi Ghane
- Modeling of Noncommunicable Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - F Mehri
- Nutrition Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
193
|
Jha NK, Sharma C, Hashiesh HM, Arunachalam S, Meeran MN, Javed H, Patil CR, Goyal SN, Ojha S. β-Caryophyllene, A Natural Dietary CB2 Receptor Selective Cannabinoid can be a Candidate to Target the Trinity of Infection, Immunity, and Inflammation in COVID-19. Front Pharmacol 2021; 12:590201. [PMID: 34054510 PMCID: PMC8163236 DOI: 10.3389/fphar.2021.590201] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 02/04/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease (COVID-19), caused by novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is an ongoing pandemic and presents a public health emergency. It has affected millions of people and continues to affect more, despite tremendous social preventive measures. Identifying candidate drugs for the prevention and treatment of COVID-19 is crucial. The pathogenesis and the complications with advanced infection mainly involve an immune-inflammatory cascade. Therefore, therapeutic strategy relies on suppressing infectivity and inflammation, along with immune modulation. One of the most promising therapeutic targets for the modulation of immune-inflammatory responses is the endocannabinoid system, particularly the activation of cannabinoid type 2 receptors (CB2R), a G-protein coupled receptor which mediates the anti-inflammatory properties by modulating numerous signaling pathways. To pharmacologically activate the CB2 receptors, a naturally occurring cannabinoid ligand, beta-caryophyllene (BCP), received attention due to its potent anti-inflammatory, antiviral, and immunomodulatory properties. BCP is recognized as a full selective functional agonist on CB2 receptors and produces therapeutic effects by activating CB2 and the nuclear receptors, peroxisome proliferator-activated receptors (PPARs). BCP is regarded as the first dietary cannabinoid with abundant presence across cannabis and non-cannabis plants, including spices and other edible plants. BCP showed tissue protective properties and favorably modulates numerous signaling pathways and inhibits inflammatory mediators, including cytokines, chemokines, adhesion molecules, prostanoids, and eicosanoids. Based on its pharmacological properties, molecular mechanisms, and the therapeutic potential of BCP as an immunomodulator, anti-inflammatory, organ-protective, and antiviral, we hypothesize that BCP could be a promising therapeutic and/or preventive candidate to target the triad of infection, immunity, and inflammation in COVID-19. In line with numerous studies that proposed the potential of cannabinoids in COVID-19, BCP may be a novel candidate compound for pharmaceutical and nutraceutical development due to its unique functional receptor selectivity, wide availability and accessibility, dietary bioavailability, nonpsychoactivity, and negligible toxicity along with druggable properties, including favorable pharmacokinetic and physicochemical properties. Based on reasonable pharmacological mechanisms and therapeutic properties, we speculate that BCP has potential to be investigated against COVID-19 and will inspire further preclinical and clinical studies.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, India
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Hebaallah Mamdouh Hashiesh
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Seenipandi Arunachalam
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mf Nagoor Meeran
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Hayate Javed
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Chandragouda R Patil
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Sameer N Goyal
- Shri Vile Parle Kelvani Mandal's Institute of Pharmacy, Dhule, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
194
|
Singh B, Mal G, Verma V, Tiwari R, Khan MI, Mohapatra RK, Mitra S, Alyami SA, Emran TB, Dhama K, Moni MA. Stem cell therapies and benefaction of somatic cell nuclear transfer cloning in COVID-19 era. Stem Cell Res Ther 2021; 12:283. [PMID: 33980321 PMCID: PMC8114669 DOI: 10.1186/s13287-021-02334-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/12/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The global health emergency of COVID-19 has necessitated the development of multiple therapeutic modalities including vaccinations, antivirals, anti-inflammatory, and cytoimmunotherapies, etc. COVID-19 patients suffer from damage to various organs and vascular structures, so they present multiple health crises. Mesenchymal stem cells (MSCs) are of interest to treat acute respiratory distress syndrome (ARDS) caused by SARS-CoV-2 infection. MAIN BODY Stem cell-based therapies have been verified for prospective benefits in copious preclinical and clinical studies. MSCs confer potential benefits to develop various cell types and organoids for studying virus-human interaction, drug testing, regenerative medicine, and immunomodulatory effects in COVID-19 patients. Apart from paving the ways to augment stem cell research and therapies, somatic cell nuclear transfer (SCNT) holds unique ability for a wide range of health applications such as patient-specific or isogenic cells for regenerative medicine and breeding transgenic animals for biomedical applications. Being a potent cell genome-reprogramming tool, the SCNT has increased prominence of recombinant therapeutics and cellular medicine in the current era of COVID-19. As SCNT is used to generate patient-specific stem cells, it avoids dependence on embryos to obtain stem cells. CONCLUSIONS The nuclear transfer cloning, being an ideal tool to generate cloned embryos, and the embryonic stem cells will boost drug testing and cellular medicine in COVID-19.
Collapse
Affiliation(s)
- Birbal Singh
- ICAR-Indian Veterinary Research Institute Regional Station, Palampur, Himachal Pradesh, India
| | - Gorakh Mal
- ICAR-Indian Veterinary Research Institute Regional Station, Palampur, Himachal Pradesh, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Muhammad Imran Khan
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, China
| | - Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, Odisha, India
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Salem A Alyami
- Department of Mathematics and Statistics, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11432, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh.
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India.
| | - Mohammad Ali Moni
- WHO Collaborating Centre on eHealth, UNSW Digital Health, Faculty of Medicine, School of Public Health and Community Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia.
| |
Collapse
|
195
|
Abstract
Die Prävalenz der venösen thromboembolischen (VTE) Ereignisse ist bei Coronavirus diesease 2019 (COVID-19) -Patienten hoch, insbesondere bei schwer Erkrankten. Patienten mit schwerer COVID-19 und VTE haben eine signifikant höhere Mortalität im Vergleich zu Patienten ohne VTE. Die Manifestation einer schweren Infektion mit Severe acute respiratory syndrome coronavirus-2 (SARS-CoV‑2) entspricht einem systemischen proinflammatorischen und prokoagulatorischen Phänotyp, der mit vaskulären Thrombosen nicht nur in den Venen, sondern auch in den Arterien, Kapillaren sowie mit einer Inflammation der Gefäße assoziiert ist. Ein erhöhter D‑Dimer-Spiegel kann als Indikator für VTE bei Patienten mit COVID-19 verwendet werden. Die meisten medizinischen Gesellschaften empfehlen eine VTE-Prophylaxe vorzugsweise mit niedermolekularen Heparinen (LMWH) bei allen stationären Patienten. Weitere Daten von randomisierten kontrollierten Studien (RCTs) über die optimale Antikoagulation und antithrombotische Therapie werden in der nahen Zukunft erwartet.
Collapse
Affiliation(s)
- Stanislava Tzaneva
- Universitätsklinik für Dermatologie, Medizinische Universität Wien, Währinger Gürtel 18–20, 1090 Wien, Österreich
| |
Collapse
|
196
|
Lin H, Wang X, Liu M, Huang M, Shen Z, Feng J, Yang H, Li Z, Gao J, Ye X. Exploring the treatment of COVID-19 with Yinqiao powder based on network pharmacology. Phytother Res 2021; 35:2651-2664. [PMID: 33452734 PMCID: PMC8013442 DOI: 10.1002/ptr.7012] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 12/18/2020] [Accepted: 12/28/2020] [Indexed: 12/24/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In China, Yinqiao powder is widely used to prevent and treat COVID-19 patients with Weifen syndrome. In this study, the screening and verification of active ingredients, target selection and DisGeNET scoring, drug-ingredient-gene network construction, protein-protein interaction network construction, molecular docking and surface plasmon resonance (SPR) analysis, gene ontology (GO) functional analysis, gene tissue analysis, and kyoto encyclopedia of genes and genomes (KEGG) pathway analysis were used to explore the active ingredients, targets, and potential mechanisms of Yinqiao powder in the treatment of COVID-19. We also predicted the therapeutic effect of Yinqiao powder using TCM anti-COVID-19 (TCMATCOV). Yinqiao powder has a certain therapeutic effect on COVID-19, with an intervention score of 20.16. Hesperetin, eriodictyol, luteolin, quercetin, and naringenin were the potentially effective active ingredients against COVID-19. The hub-proteins were interleukin-6 (IL-6), mitogen-activated protein kinase 3 (MAPK3), tumor necrosis factor (TNF), and tumor protein P53 (TP53). The potential mechanisms of Yinqiao powder in the treatment of COVID-19 are the TNF signaling pathway, T-cell receptor signaling pathway, Toll-like receptor signaling pathway, and MAPK signaling pathway. This study provides a new perspective for discovering potential drugs and mechanisms of COVID-19.
Collapse
Affiliation(s)
- Haixiong Lin
- Lingnan Medical Research CenterThe First School of Clinical Medicine, Guangzhou University of Chinese MedicineGuangzhouChina
| | - Xiaotong Wang
- South China Research Center for Acupuncture and MoxibustionClinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese MedicineGuangzhouChina
| | - Minyi Liu
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Minling Huang
- Lingnan Medical Research CenterThe First School of Clinical Medicine, Guangzhou University of Chinese MedicineGuangzhouChina
| | - Zhen Shen
- Department of OrthopaedicsKunming Municipal Hospital of Traditional Chinese Medicine, The Third Affiliated Hospital of Yunnan University of Chinese MedicineKunmingChina
| | - Junjie Feng
- Lingnan Medical Research CenterThe First School of Clinical Medicine, Guangzhou University of Chinese MedicineGuangzhouChina
| | - Huijun Yang
- Department of Rehabilitation MedicineThe Sixth School of Clinical Medicine & Shenzhen Hospital, Guangzhou University of Chinese MedicineShenzhenChina
| | - Zige Li
- Lingnan Medical Research CenterThe First School of Clinical Medicine, Guangzhou University of Chinese MedicineGuangzhouChina
| | - Junyan Gao
- Lingnan Medical Research CenterThe First School of Clinical Medicine, Guangzhou University of Chinese MedicineGuangzhouChina
| | - Xiaopeng Ye
- Department of GastroenterologyShenzhen Bao'an Traditional Chinese Medicine Hospital Group, Guangzhou University of Chinese MedicineShenzhenChina
| |
Collapse
|
197
|
Corrao S, Mallaci Bocchio R, Lo Monaco M, Natoli G, Cavezzi A, Troiani E, Argano C. Does Evidence Exist to Blunt Inflammatory Response by Nutraceutical Supplementation during COVID-19 Pandemic? An Overview of Systematic Reviews of Vitamin D, Vitamin C, Melatonin, and Zinc. Nutrients 2021; 13:1261. [PMID: 33921297 PMCID: PMC8069903 DOI: 10.3390/nu13041261] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 02/07/2023] Open
Abstract
More than one year has passed since the first cases of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome (SARS)-CoV-2 coronavirus were reported in Wuhan (China), rapidly evolving into a global pandemic. This infectious disease has become a major public health challenge in the world. Unfortunately, to date, no specific antivirals have been proven to be effective against COVID-19, and although a few vaccines are available, the mortality rate is not decreasing but is still increasing. One therapeutic strategy has been focused on infection prevention and control measures. In this regard, the use of nutraceutical supports may play a role against some aspect of the infection, particularly the inflammatory state and the immune system function of patients, thus representing a strategy to control the worst outcomes of this pandemic. For this reason, we performed an overview including meta-analyses and systematic reviews to assess the association among melatonin, vitamin C, vitamin D, zinc supplementation and inflammatory markers using three databases, namely, MEDLINE, PubMed Central and the Cochrane Library of Systematic Reviews. According to the evidence available, an intake of 50,000 IU/month of vitamin D showed efficacy in CRP. An amount of 1 to 2 g per day of vitamin C demonstrated efficacy both in CRP and endothelial function, and a dosage of melatonin ranging from 5 to 25 mg /day showed good evidence of efficacy in CRP, TNF and IL6. A dose of 50 mg/day of elemental zinc supplementation showed positive results in CRP. Based on the data reported in this review, the public health system could consider whether it is possible to supplement the current limited preventive measures through targeted nutraceutical large-scale administration.
Collapse
Affiliation(s)
- Salvatore Corrao
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, [PROMISE], University of Palermo, 90127 Palermo, Italy
- COVID Unit, Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (R.M.B.); (M.L.M.); (G.N.); (C.A.)
| | - Raffaella Mallaci Bocchio
- COVID Unit, Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (R.M.B.); (M.L.M.); (G.N.); (C.A.)
| | - Marika Lo Monaco
- COVID Unit, Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (R.M.B.); (M.L.M.); (G.N.); (C.A.)
| | - Giuseppe Natoli
- COVID Unit, Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (R.M.B.); (M.L.M.); (G.N.); (C.A.)
| | - Attilio Cavezzi
- Eurocenter Venalinfa, 63074 San Benedetto del Tronto, Italy;
| | - Emidio Troiani
- Cardiology Unit, State Hospital, Social Security Institute, 20, 47893 Cailungo, San Marino;
| | - Christiano Argano
- COVID Unit, Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; (R.M.B.); (M.L.M.); (G.N.); (C.A.)
| |
Collapse
|
198
|
Cao Q, Lei H, Yang M, Wei L, Dong Y, Xu J, Nasser M, Liu M, Zhu P, Xu L, Zhao M. Impact of Cardiovascular Diseases on COVID-19: A Systematic Review. Med Sci Monit 2021; 27:e930032. [PMID: 33820904 PMCID: PMC8035813 DOI: 10.12659/msm.930032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In December 2019, pneumonia of unknown cause broke out, and currently more than 150 countries around the world have been affected. Globally, as of 5: 46 pm CET, 6 November 2020, the World Health Organization (WHO) had reported 48 534 508 confirmed cases of COVID-19, including 1 231 017 deaths. The novel coronavirus disease (COVID-19) outbreak, caused by the SARS-CoV-2 virus, is the most important medical challenge in decades. Previous research mainly focused on the exploration of lung changes. However, with development of the disease and deepening research, more and more patients showed cardiovascular diseases, even in those without respiratory symptoms, and some researchers have found that underlying cardiovascular diseases increase the risk of infection. Although the related mechanism is not thoroughly studied, based on existing research, we speculate that the interaction between the virus and its receptor, inflammatory factors, various forms of the stress response, hypoxic environment, and drug administration could all induce the development of cardiac adverse events. Interventions to control these pathogenic factors may effectively reduce the occurrence of cardiovascular complications. This review summarizes the latest research on the relationship between COVID-19 and its associated cardiovascular complications, and we also explore possible mechanisms and treatments.
Collapse
Affiliation(s)
- Qingtai Cao
- Hunan Normal University School of Medicine, Changsha, Hunan, China (mainland)
| | - HanYu Lei
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - MengLing Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Le Wei
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - YinMiao Dong
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - JiaHao Xu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Mi Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China (mainland)
| | - MengQi Liu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China (mainland)
| | - LinYong Xu
- School of Life Science, Central South University, Changsha, Hunan, China (mainland)
| | - MingYi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
199
|
Tanwar O, Soni A, Prajapat P, Shivhare T, Pandey P, Samaiya PK, Pandey SP, Kar P. Ethyl Pyruvate as a Potential Defense Intervention against Cytokine Storm in COVID-19? ACS OMEGA 2021; 6:7754-7760. [PMID: 33778286 PMCID: PMC7992141 DOI: 10.1021/acsomega.1c00157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/23/2021] [Indexed: 06/12/2023]
Abstract
COVID-19 is a deadly pandemic and has resulted in a huge loss of money and life in the past few months. It is well known that the SARS-CoV-2 gene mutates relatively slowly as compared to other viruses but still may create hurdles in developing vaccines. Therefore, there is a need to develop alternative routes for its management and treatment of COVID-19. Based on the severity of viral infection in COVID-19 patients, critically ill patients (∼5%, with old age, and comorbidities) are at high risk of morbidities. The reason for this severity in such patients is attributed to "misleading cytokine storm", which produces ARDS and results in the deaths of critically ill patients. In this connection, ethyl pyruvate (EP) controls these cytokines/chemokines, is an anti-inflammatory agent, and possesses a protective effect on the lungs, brain, heart, and mitochondria against various injuries. Considering these facts, we propose that the site-selective EP formulations (especially aerosols) could be the ultimate adjuvant therapy for the regulation of misleading cytokine storm in severely affected COVID-19 patients and could reduce the mortalities.
Collapse
Affiliation(s)
- Omprakash Tanwar
- Department
of Pharmacy, Shri G.S. Institute of Technology
and Science, 23, Sir
M. Visvesvaraya Marg (Park Road), Indore 452003, Madhya Pradesh, India
| | - Aastha Soni
- Department
of Pharmacy, Shri G.S. Institute of Technology
and Science, 23, Sir
M. Visvesvaraya Marg (Park Road), Indore 452003, Madhya Pradesh, India
| | - Pawan Prajapat
- Department
of Pharmacy, Shri G.S. Institute of Technology
and Science, 23, Sir
M. Visvesvaraya Marg (Park Road), Indore 452003, Madhya Pradesh, India
| | - Tanu Shivhare
- Department
of Pharmacy, Shri G.S. Institute of Technology
and Science, 23, Sir
M. Visvesvaraya Marg (Park Road), Indore 452003, Madhya Pradesh, India
| | - Pooja Pandey
- Department
of Pharmacy, Shri G.S. Institute of Technology
and Science, 23, Sir
M. Visvesvaraya Marg (Park Road), Indore 452003, Madhya Pradesh, India
| | - Puneet Kumar Samaiya
- Department
of Pharmacy, Shri G.S. Institute of Technology
and Science, 23, Sir
M. Visvesvaraya Marg (Park Road), Indore 452003, Madhya Pradesh, India
| | - Sharad Prakash Pandey
- Department
of Pharmacy, Shri G.S. Institute of Technology
and Science, 23, Sir
M. Visvesvaraya Marg (Park Road), Indore 452003, Madhya Pradesh, India
| | - Parimal Kar
- Department
of Biosciences & Biomedical Engineering, Indian Institute of Technology, Khandwa Road, Simrol, Indore 453552, Madhya Pradesh, India
| |
Collapse
|
200
|
Perfetto L, Micarelli E, Iannuccelli M, Lo Surdo P, Giuliani G, Latini S, Pugliese GM, Massacci G, Vumbaca S, Riccio F, Fuoco C, Paoluzi S, Castagnoli L, Cesareni G, Licata L, Sacco F. A Resource for the Network Representation of Cell Perturbations Caused by SARS-CoV-2 Infection. Genes (Basel) 2021; 12:450. [PMID: 33809949 PMCID: PMC8004236 DOI: 10.3390/genes12030450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has caused more than 2.3 million casualties worldwide and the lack of effective treatments is a major health concern. The development of targeted drugs is held back due to a limited understanding of the molecular mechanisms underlying the perturbation of cell physiology observed after viral infection. Recently, several approaches, aimed at identifying cellular proteins that may contribute to COVID-19 pathology, have been reported. Albeit valuable, this information offers limited mechanistic insight as these efforts have produced long lists of cellular proteins, the majority of which are not annotated to any cellular pathway. We have embarked in a project aimed at bridging this mechanistic gap by developing a new bioinformatic approach to estimate the functional distance between a subset of proteins and a list of pathways. A comprehensive literature search allowed us to annotate, in the SIGNOR 2.0 resource, causal information underlying the main molecular mechanisms through which severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and related coronaviruses affect the host-cell physiology. Next, we developed a new strategy that enabled us to link SARS-CoV-2 interacting proteins to cellular phenotypes via paths of causal relationships. Remarkably, the extensive information about inhibitors of signaling proteins annotated in SIGNOR 2.0 makes it possible to formulate new potential therapeutic strategies. The proposed approach, which is generally applicable, generated a literature-based causal network that can be used as a framework to formulate informed mechanistic hypotheses on COVID-19 etiology and pathology.
Collapse
Affiliation(s)
- Livia Perfetto
- Fondazione Human Technopole, Department of Biology, Via Cristina Belgioioso, 171, 20157 Milan, Italy; (L.P.); (P.L.S.)
| | - Elisa Micarelli
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Marta Iannuccelli
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Prisca Lo Surdo
- Fondazione Human Technopole, Department of Biology, Via Cristina Belgioioso, 171, 20157 Milan, Italy; (L.P.); (P.L.S.)
| | - Giulio Giuliani
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Sara Latini
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Giusj Monia Pugliese
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Giorgia Massacci
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Simone Vumbaca
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Federica Riccio
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Serena Paoluzi
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Luana Licata
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (E.M.); (M.I.); (G.G.); (S.L.); (G.M.P.); (G.M.); (S.V.); (F.R.); (C.F.); (S.P.); (L.C.); (G.C.); (L.L.)
| |
Collapse
|