1
|
Alirezaee A, Mirmoghtadaei M, Heydarlou H, Akbarian A, Alizadeh Z. Interferon therapy in alpha and Delta variants of SARS-CoV-2: The dichotomy between laboratory success and clinical realities. Cytokine 2024; 186:156829. [PMID: 39693873 DOI: 10.1016/j.cyto.2024.156829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
The COVID-19 pandemic has caused significant morbidity and mortality worldwide. The emergence of the Alpha and Delta variants of SARS-CoV-2 has led to a renewed interest in using interferon therapy as a potential treatment option. Interferons are a group of signaling proteins produced by host cells in response to viral infections. They play a critical role in the innate immune response to viral infections by inducing an antiviral state in infected and neighboring cells. Interferon therapy has shown promise as a potential treatment option for COVID-19. In this review paper, we review the current knowledge regarding interferon therapy in the context of the Alpha and Delta variants of SARS-CoV-2 and discuss the challenges that must be overcome to translate laboratory findings into effective clinical treatments.
Collapse
Affiliation(s)
- Atefe Alirezaee
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Mirmoghtadaei
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh Heydarlou
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Asiye Akbarian
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Alizadeh
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Trimarco V, Izzo R, Pacella D, Virginia Manzi M, Trama U, Lembo M, Piccinocchi R, Gallo P, Esposito G, Morisco C, Rozza F, Mone P, Jankauskas SS, Piccinocchi G, Santulli G, Trimarco B. Increased prevalence of cardiovascular-kidney-metabolic syndrome during COVID-19: A propensity score-matched study. Diabetes Res Clin Pract 2024; 218:111926. [PMID: 39536978 DOI: 10.1016/j.diabres.2024.111926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
A recent presidential advisory from the American Heart Association (AHA) has introduced the term cardiovascular-kidney-metabolic (CKM) syndrome to describe the complex interplay among health conditions linking heart, kidney, and metabolism. The aim of our study was to compare the prevalence of concurrent CKM syndrome components before and during the COVID-19 pandemic and identify associated risk factors. We conducted a study utilizing data from a real-world population obtained from a primary care database. The study cohort comprised a closed group followed over a 6-year period (2017-2022). A total of 81,051 individuals were included: 32,650 in the pre-pandemic period and 48,401 in the 2020-2022 triennium. After propensity-score matching for sex, age, and BMI, the study included 30,511 participants for each period. 3554 individuals were diagnosed with type 2 diabetes in the pre-pandemic period, compared to 7430 during the pandemic. Hypertension, dyslipidemia, and obesity displayed significant increases in prevalence during the pandemic, and prediabetes had a particularly sharp rise of 170%. Age-stratified analyses revealed a higher burden of CKM conditions with advancing age. Our findings indicate a substantial increase in the prevalence of CKM syndrome during the COVID-19 pandemic, with nearly half of the patients exhibiting one or more CKM syndrome components.
Collapse
Affiliation(s)
- Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences, and Dentistry, "Federico II" University, Naples, Italy
| | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Daniela Pacella
- Department of Public Health, "Federico II" University, Naples, Italy
| | - Maria Virginia Manzi
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Ugo Trama
- Pharmaceutical Department of Campania Region, Naples, Italy
| | - Maria Lembo
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | | | - Paola Gallo
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Carmine Morisco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy; International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy; Italian Society for Cardiovascular Prevention (SIPREC), Rome, Italy
| | - Francesco Rozza
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Pasquale Mone
- Department of Medicine and Health Sciences "Vincenzo Tiberio", Molise University, Campobasso, Italy; Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, The Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY, USA; Casa di Cura "Montevergine", Mercogliano (Avellino), Italy
| | - Stanislovas S Jankauskas
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, The Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY, USA
| | - Gaetano Piccinocchi
- COMEGEN Primary Care Physicians Cooperative, Italian Society of General Medicine (SIMG), Naples, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy; International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy; Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, The Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY, USA; Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York City, NY, USA.
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy; International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy; Italian Society for Cardiovascular Prevention (SIPREC), Rome, Italy
| |
Collapse
|
3
|
Alotaibi B, Elekhnawy E, El-Masry TA, Saleh A, Alosaimi ME, Alotaibi KN, Negm WA. Antibacterial potential of Euphorbia canariensis against Pseudomonas aeruginosa bacteria causing respiratory tract infections. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:261-269. [PMID: 38696143 DOI: 10.1080/21691401.2024.2345891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 04/14/2024] [Indexed: 05/14/2024]
Abstract
The widespread dissemination of bacterial resistance has led to great attention being paid to finding substitutes for traditionally used antibiotics. Plants are rich in various phytochemicals that could be used as antibacterial therapies. Here, we elucidate the phytochemical profile of Euphorbia canariensis ethanol extract (EMEE) and then elucidate the antibacterial potential of ECEE against Pseudomonas aeruginosa clinical isolates. ECEE showed minimum inhibitory concentrations ranging from 128 to 512 µg/mL. The impact of ECEE on the biofilm-forming ability of the tested isolates was elucidated using crystal violet assay and qRT-PCR to study its effect on the gene expression level. ECEE exhibited antibiofilm potential, which resulted in a downregulation of the expression of the biofilm genes (algD, pelF, and pslD) in 39.13% of the tested isolates. The antibacterial potential of ECEE was studied in vivo using a lung infection model in mice. A remarkable improvement was observed in the ECEE-treated group, as revealed by the histological and immunohistochemical studies. Also, ELISA showed a noticeable decrease in the oxidative stress markers (nitric oxide and malondialdehyde). The gene expression of the proinflammatory marker (interleukin-6) was downregulated, while the anti-inflammatory biomarker was upregulated (interleukin-10). Thus, clinical trials should be performed soon to explore the potential antibacterial activity of ECEE, which could help in our battle against resistant pathogenic bacteria.
Collapse
Affiliation(s)
- Badriyah Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Manal E Alosaimi
- Department of Basic Health Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | | | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta
| |
Collapse
|
4
|
Abe S, Wannigama DL, Suzuki Y, Akaneya D, Igarashi J, Suto M, Moriya K, Ishizawa D, Okuma Y, Hongsing P, Hurst C, Saethang T, Higgins PG, Stick SM, Kicic A. Real world effectiveness of early ensitrelvir treatment in patients with SARS-CoV-2, a retrospective case series. New Microbes New Infect 2024; 62:101522. [PMID: 39552926 PMCID: PMC11567130 DOI: 10.1016/j.nmni.2024.101522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
Background Ensitrelvir, a 3C-like protease inhibitor, received emergency approval in Japan in November 2022 for treating non-hospitalized patients with mild-to-moderate COVID-19. However, confirmation of its real-world clinical effectiveness is limited. Methods This retrospective study evaluated 18 vaccinated outpatients (15 men; median age, 39.5 years; range, 26-56), treated with a 5-day oral ensitrelvir regimen (375 mg loading dose, followed by 125 mg daily) between December 1, 2022, and January 31, 2023. Nasal swabs were collected on days 0, 3, 6, and 9 for RT-qPCR to assess viral load. Variants were identified by Sanger sequencing, and outcomes were compared to historical controls. Patients were followed for 60 days to monitor for post-acute sequelae of COVID-19 (PASC). Results Symptoms such as mild fever and sore throat improved rapidly after one day of ensitrelvir treatment, with 66 % of patients recovering within six days. All individuals were infected with the BA.5 Omicron variant. Viral loads, as measured by Ct values, increased significantly from 21.82 at symptom onset to 37.65 b y day 6, with SARS-CoV-2 RNA undetectable in most patients by day 9. Those treated within 48 h of symptom onset showed the viral load reduction. Compared to historical controls, where symptom resolution took 8.5 days, ensitrelvir shortened recovery time to as little as 1.4 days for over 66 % of patients. Conclusion Ensitrelvir treatment resulted in rapid symptom relief and significant viral load reduction, with no adverse events, viral rebound, or PASC symptoms, demonstrating its potential efficacy and safety. Larger studies are needed for further confirmation.
Collapse
Affiliation(s)
- Shuichi Abe
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Pathogen Hunter's Research Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Pathogen Hunter's Research Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Department of Infectious Diseases, Faculty of Medicine Yamagata University, Yamagata, Japan
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
- Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield, United Kingdom
| | - Yu Suzuki
- Department of Clinical Laboratory, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Daisuke Akaneya
- Department of Clinical Laboratory, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Junko Igarashi
- Department of Clinical Laboratory, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Mayu Suto
- Department of Clinical Laboratory, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Kazunori Moriya
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Daisuke Ishizawa
- Department of Pharmacy, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Yoshikazu Okuma
- Department of Pharmacy, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Parichart Hongsing
- Mae Fah Luang University Hospital, Chiang Rai, Thailand
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Cameron Hurst
- Molly Wardaguga Research Centre, Charles Darwin University, Queensland, Australia
- Department of Clinical Epidemiology, Faculty of Medicine, Thammasat University, Rangsit, Thailand, 10120
- Center of Excellence in Applied Epidemiology, Thammasat University, Rangsit, Thailand, 10120
| | - Thammakorn Saethang
- Department of Computer Science, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Paul G. Higgins
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - Stephen M. Stick
- Wal-yan Respiratory Centre, Telethon Kids Institute, University of Western Australia, Nedlands, 6009, Western Australia, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, 6009, Western Australia, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Centre, Telethon Kids Institute, University of Western Australia, Nedlands, 6009, Western Australia, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, 6009, Western Australia, Australia
- School of Population Health, Curtin University, Bentley, 6102, Western Australia, Australia
| |
Collapse
|
5
|
Kurmanova G, Zhanaev A, Kaldybek A, Abdrakhmanova B, Akparova A. Impact of the COVID-19 pandemic on the clinical features of patients with chronic obstructive pulmonary disease: an observational cross-sectional study. Monaldi Arch Chest Dis 2024. [PMID: 39569839 DOI: 10.4081/monaldi.2024.3128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024] Open
Abstract
The presence of chronic obstructive pulmonary disease (COPD) and COVID-19 infection is a detrimental combination for patients and can cause negative clinical consequences. The investigation aimed to compare sociodemographic and clinical parameters of COPD individuals hospitalized for exacerbations before and at the end of the COVID-19 pandemic. An observational cross-sectional study including 222 patients with COPD was conducted in two stages: a survey and assessment of clinical and laboratory data of patients hospitalized from September 2022 to March 2023 (n=98) and processing of the medical histories of patients with COPD who received hospital treatment in 2017 and 2018 (n=124). A comparative analysis of patients who received inpatient treatment for COPD showed that the frequency of patients with Global Initiative for Chronic Obstructive Lung Disease (GOLD) I was half as high after the COVID-19 pandemic, whereas the individuals with GOLD IV were more frequent during the same period (p<0.05). Multiple regression analysis proved the effects of smoking status and previous COVID-19 infection on the health status of patients with COPD according to COPD Assessment Test data (p<0.05). There was an increase in the frequency of comorbid pathologies in the post-COVID period: hypertension, coronary heart disease, gastrointestinal diseases, anemia (p<0.05), and other diseases. This study highlights the significant influence of the COVID-19 infection on people with COPD, which manifested as impaired lung function and an increased incidence of comorbidities.
Collapse
Affiliation(s)
- Gaukhar Kurmanova
- Department of Clinical Subjects, Al-Farabi Kazakh National University, Almaty
| | - Almas Zhanaev
- Department of Pulmonology, City Clinical Hospital No. 1, Almaty
| | | | - Balkiya Abdrakhmanova
- Department of General Biology and Genomics, L.N. Gumilyov Eurasian National University, Astana
| | - Almira Akparova
- Department of Clinical Subjects, Al-Farabi Kazakh National University, Almaty
| |
Collapse
|
6
|
Wendt R, Macholz M, Kalbitz S, Herrmann N, Herbst V, Hammes T, Kai M, Ankersmit HJ, Beige J, Lübbert C, Graf A, Scherberich J. Serum uromodulin associates with kidney function and outcome in a cohort of hospitalised COVID-19 patients. Sci Rep 2024; 14:25420. [PMID: 39455668 PMCID: PMC11512065 DOI: 10.1038/s41598-024-76372-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the prevalence and evaluates the prognostic implications of acute kidney injury (AKI) in COVID-19 patients, with a novel emphasis on the evaluation of serum uromodulin (sUmod) as a potential kidney-specific biomarker. A cohort of hospitalised COVID-19 patients (n = 378) was examined for AKI using standard criteria. In addition to traditional urinary biomarkers, sUmod levels were analysed. Univariable and multivariable regression models were employed to evaluate the association of sUmod and AKI and in-hospital mortality. Levels of sUmod were significantly lower in patients with CKD (91.8 ± 60.7 ng/ml) compared to patients with normal kidney function (204.7 ± 91.7 ng/ml; p < 0.001). 151 patients (40.0%) presented with AKI at the time of hospital admission or developed an AKI during hospitalization. 116 patients (76.8%) had an AKI already at the time of hospital admission. COVID-19 patients with AKI had significantly lower levels of sUmod compared to patients without AKI during hospitalisation (124.8 ± 79.5 ng/ml) vs 214.6 ± 92.3 ng/ml; p < 0.001). The in-hospital mortality rate in this cohort of COVID-19 patients was 15.3%. Patients with AKI had a higher probability for in-hospital death (OR 5.6, CI 1.76 to 17.881, p = 0.004). Patients who died during hospital stay, had significantly lower sUmod levels (129.14 ± 89.56 ng/ml) compared to patients surviving hospitalisation (187.71 ± 96,64 ng/ml; p < 0.001). AKI is frequently associated with COVID-19 in hospitalized patients. Serum uromodulin may emerge as a promising biomarker for AKI in COVID-19 patients. Further research is warranted to explore its clinical application and refine risk stratification in this patient population.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Nephrology, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany.
| | - Martin Macholz
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Nadja Herrmann
- Department of Finance and Controlling, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Victor Herbst
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Tabea Hammes
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Marco Kai
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Hendrik Jan Ankersmit
- Clinic of Thoracic Surgery, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Joachim Beige
- Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, Delitzscher Str. 141, 04129, Leipzig, Germany
- Martin-Luther University Halle/Wittenberg, Halle, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Leipzig University Hospital, Liebigstr. 20, 04103, Leipzig, Germany
| | - Alexandra Graf
- Institute of Medical Statistics, Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Jürgen Scherberich
- Department of Nephrology and Clinical Immunology, Klinikum München, 81545, Munich, Germany
| |
Collapse
|
7
|
Liu Z, Petinrin OO, Chen N, Toseef M, Liu F, Zhu Z, Qi F, Wong KC. Identification and evaluation of candidate COVID-19 critical genes and medicinal drugs related to plasma cells. BMC Infect Dis 2024; 24:1099. [PMID: 39363208 PMCID: PMC11451256 DOI: 10.1186/s12879-024-10000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
The ongoing COVID-19 pandemic, caused by the SARS-CoV-2 virus, represents one of the most significant global health crises in recent history. Despite extensive research into the immune mechanisms and therapeutic options for COVID-19, there remains a paucity of studies focusing on plasma cells. In this study, we utilized the DESeq2 package to identify differentially expressed genes (DEGs) between COVID-19 patients and controls using datasets GSE157103 and GSE152641. We employed the xCell algorithm to perform immune infiltration analyses, revealing notably elevated levels of plasma cells in COVID-19 patients compared to healthy individuals. Subsequently, we applied the Weighted Gene Co-expression Network Analysis (WGCNA) algorithm to identify COVID-19 related plasma cell module genes. Further, positive cluster biomarker genes for plasma cells were extracted from single-cell RNA sequencing data (GSE171524), leading to the identification of 122 shared genes implicated in critical biological processes such as cell cycle regulation and viral infection pathways. We constructed a robust protein-protein interaction (PPI) network comprising 89 genes using Cytoscape, and identified 20 hub genes through cytoHubba. These genes were validated in external datasets (GSE152418 and GSE179627). Additionally, we identified three potential small molecules (GSK-1070916, BRD-K89997465, and idarubicin) that target key hub genes in the network, suggesting a novel therapeutic approach. These compounds were characterized by their ability to down-regulate AURKB, KIF11, and TOP2A effectively, as evidenced by their low free binding energies determined through computational analyses using cMAP and AutoDock. This study marks the first comprehensive exploration of plasma cells' role in COVID-19, offering new insights and potential therapeutic targets. It underscores the importance of a systematic approach to understanding and treating COVID-19, expanding the current body of knowledge and providing a foundation for future research.
Collapse
Affiliation(s)
- Zhe Liu
- Institute for Hepatology, The Second Affiliated Hospital, School of Medicine, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518112, China
- Department of Computer Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | | | - Nanjun Chen
- Department of Computer Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Muhammad Toseef
- Department of Computer Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fang Liu
- Rocgene (Beijing) Technology Co., Ltd, Beijing, Beijing, 102200, China
| | - Zhongxu Zhu
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
| | - Furong Qi
- Institute for Hepatology, The Second Affiliated Hospital, School of Medicine, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518112, China.
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
8
|
Mariappan V, Adla D, Jangili S, Ranganadin P, Green SR, Mohammed S, Mutheneni SR, Pillai AB. Understanding COVID-19 outcome: Exploring the prognostic value of soluble biomarkers indicative of endothelial impairment. Cytokine 2024; 180:156673. [PMID: 38857562 DOI: 10.1016/j.cyto.2024.156673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
Host proteins released by the activated endothelial cells during SARS-CoV-2 infection are implicated to be involved in coagulation and endothelial dysfunction. However, the underlying mechanism that governs the vascular dysfunction and disease severity in COVID-19 remains obscure. The study evaluated the serum levels of Bradykinin, Kallikrein, SERPIN A, and IL-18 in COVID-19 (N-42 with 20 moderate and 22 severe) patients compared to healthy controls (HC: N-10) using ELISA at the day of admission (DOA) and day 7 post-admission. The efficacy of the protein levels in predicting disease severity was further determined using machine learning models. The levels of bradykinins and SERPIN A were higher (P ≤ 0.001) in both severe and moderate cases on day 7 post-admission compared to DOA. All the soluble proteins studied were found to elevated (P ≤ 0.01) in severe compared to moderate in day 7 and were positively correlated (P ≤ 0.001) with D-dimer, a marker for coagulation. ROC analysis identified that SERPIN A, IL-18, and bradykinin could predict the clinical condition of COVID-19 with AUC values of 1, 0.979, and 1, respectively. Among the models trained using univariate model analysis, SERPIN A emerged as a strong prognostic biomarker for COVID-19 disease severity. The serum levels of SERPIN A in conjunction with the coagulation marker D-dimer, serve as a predictive indicator for COVID-19 clinical outcomes. However, studies are required to ascertain the role of these markers in disease virulence.
Collapse
Affiliation(s)
- Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Deepthi Adla
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Shraddha Jangili
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Pajanivel Ranganadin
- Department of Pulmonary Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Siva Ranaganthan Green
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Salma Mohammed
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Srinivasa Rao Mutheneni
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| |
Collapse
|
9
|
Di Spigna G, Covelli B, Vargas M, Di Caprio R, Rubino V, Iacovazzo C, Napolitano F, Servillo G, Postiglione L. The Behaviour of IL-6 and Its Soluble Receptor Complex during Different Waves of the COVID-19 Pandemic. Life (Basel) 2024; 14:814. [PMID: 39063569 PMCID: PMC11278279 DOI: 10.3390/life14070814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
In late December 2019, SARS-CoV-2 was identified as the cause of a new pneumonia (COVID-19), leading to a global pandemic declared by the WHO on 11 March 2020, with significant human, economic, and social costs. Although most COVID-19 cases are asymptomatic or mild, 14% progress to severe disease, and 5% develop critical illness with complications such as interstitial pneumonia, acute respiratory distress syndrome (ARDS), and multiple organ dysfunction syndrome (MODS). SARS-CoV-2 primarily targets the respiratory system but can affect multiple organs due to the widespread presence of angiotensin-converting enzyme 2 (ACE2) receptors, which the virus uses to enter cells. This broad distribution of ACE2 receptors means that SARS-CoV-2 infection can lead to cardiovascular, gastrointestinal, renal, hepatic, central nervous system, and ocular damage. The virus triggers the innate and adaptive immune systems, resulting in a massive cytokine release, known as a "cytokine storm", which is linked to tissue damage and poor outcomes in severe lung disease. Interleukin-6 (IL-6) is particularly important in this cytokine release, with elevated levels serving as a marker of severe COVID-19. IL-6 is a multifunctional cytokine with both anti-inflammatory and pro-inflammatory properties, acting through two main pathways: classical signalling and trans-signalling. Classical signalling involves IL-6 binding to its membrane-bound receptor IL-6R and then to the gp130 protein, while trans-signalling occurs when IL-6 binds to the soluble form of IL-6R (sIL-6R) and then to membrane-bound gp130 on cells that do not express IL-6R. The soluble form of gp130 (sgp130) can inhibit IL-6 trans-signalling by binding to sIL-6R, thereby preventing it from interacting with membrane-bound gp130. Given the central role of IL-6 in COVID-19 inflammation and its association with severe disease, we aimed to analyse the behaviour of IL-6 and its soluble receptor complex during different waves of the pandemic. This analysis could help determine whether IL-6 levels can serve as prognostic markers of disease severity.
Collapse
Affiliation(s)
- Gaetano Di Spigna
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Bianca Covelli
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Maria Vargas
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.); (G.S.)
| | - Roberta Di Caprio
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Valentina Rubino
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Carmine Iacovazzo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.); (G.S.)
| | - Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Giuseppe Servillo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.); (G.S.)
| | - Loredana Postiglione
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| |
Collapse
|
10
|
de Souza SP, Caldas JR, Lopes MB, Duarte Silveira MA, Coelho FO, Oliveira Queiroz I, Domingues Cury P, Passos RDH. Physico-chemical characterization of acid base disorders in patients with COVID-19: A cohort study. World J Nephrol 2024; 13:92498. [PMID: 38983762 PMCID: PMC11229835 DOI: 10.5527/wjn.v13.i2.92498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Acid-base imbalance has been poorly described in patients with coronavirus disease 2019 (COVID-19). Study by the quantitative acid-base approach may be able to account for minor changes in ion distribution that may have been overlooked using traditional acid-base analysis techniques. In a cohort of critically ill COVID-19 patients, we looked for an association between metabolic acidosis surrogates and worse clinical outcomes, such as mortality, renal dialysis, and length of hospital stay. AIM To describe the acid-base disorders of critically ill COVID-19 patients using Stewart's approach, associating its variables with poor outcomes. METHODS This study pertained to a retrospective cohort comprised of adult patients who experienced an intensive care unit stay exceeding 4 days and who were diagnosed with severe acute respiratory syndrome coronavirus 2 infection through a positive polymerase chain reaction analysis of a nasal swab and typical pulmonary involvement observed in chest computed tomography scan. Laboratory and clinical data were obtained from electronic records. Categorical variables were compared using Fisher's exact test. Continuous data were presented as median and interquartile range. The Mann-Whitney U test was used for comparisons. RESULTS In total, 211 patients were analyzed. The mortality rate was 13.7%. Overall, 149 patients (70.6%) presented with alkalosis, 28 patients (13.3%) had acidosis, and the remaining 34 patients (16.2%) had a normal arterial pondus hydrogenii. Of those presenting with acidosis, most had a low apparent strong ion difference (SID) (20 patients, 9.5%). Within the group with alkalosis, 128 patients (61.0%) had respiratory origin. The non-survivors were older, had more comorbidities, and had higher Charlson's and simplified acute physiology score 3. We did not find severe acid-base imbalance in this population. The analyzed Stewart's variables (effective SID, apparent SID, and strong ion gap and the effect of albumin, lactate, phosphorus, and chloride) were not different between the groups. CONCLUSION Alkalemia is prevalent in COVID-19 patients. Although we did not find an association between acid-base variables and mortality, the use of Stewart's methodology may provide insights into this severe disease.
Collapse
Affiliation(s)
- Sergio Pinto de Souza
- Department of Nephrology, Hospital São Rafael, Salvador, BA 41253190, Brazil
- Department of Nephrology, D’Or Institute for Research and Education (IDOR), Salvador, BA 41253190, Brazil
- Faculty of Medicine, Escola Bahiana de Medicina e Saúde Pública-EBMSP, Salvador, BA 40290000, Brazil
| | - Juliana R Caldas
- Department of Intensive Care, D’Or Institute for Research and Education (IDOR), Salvador, BA 41253190, Brazil
| | - Marcelo Barreto Lopes
- Department of Nephrology, Hospital São Rafael, Salvador, BA 41253190, Brazil
- Department of Nephrology, D’Or Institute for Research and Education (IDOR), Salvador, BA 41253190, Brazil
| | - Marcelo Augusto Duarte Silveira
- Department of Nephrology, Hospital São Rafael, Salvador, BA 41253190, Brazil
- Department of Nephrology, D’Or Institute for Research and Education (IDOR), Salvador, BA 41253190, Brazil
| | - Fernanda Oliveira Coelho
- Department of Nephrology, Hospital São Rafael, Salvador, BA 41253190, Brazil
- Department of Nephrology, D’Or Institute for Research and Education (IDOR), Salvador, BA 41253190, Brazil
| | - Igor Oliveira Queiroz
- Hospital São Rafael, D’Or Institute for Research and Education (IDOR), Salvador, BA 41253190, Brazil
| | - Pedro Domingues Cury
- Hospital São Rafael, D’Or Institute for Research and Education (IDOR), Salvador, BA 41253190, Brazil
| | - Rogério da Hora Passos
- Department of Intensive Care Unit, Hospital Israelita Albert Einstein, Sao Paulo, SP 05652900, Brazil
| |
Collapse
|
11
|
Padilla‐Flores T, Sampieri A, Vaca L. Incidence and management of the main serious adverse events reported after COVID-19 vaccination. Pharmacol Res Perspect 2024; 12:e1224. [PMID: 38864106 PMCID: PMC11167235 DOI: 10.1002/prp2.1224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/27/2024] [Indexed: 06/13/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2n first appeared in Wuhan, China in 2019. Soon after, it was declared a pandemic by the World Health Organization. The health crisis imposed by a new virus and its rapid spread worldwide prompted the fast development of vaccines. For the first time in human history, two vaccines based on recombinant genetic material technology were approved for human use. These mRNA vaccines were applied in massive immunization programs around the world, followed by other vaccines based on more traditional approaches. Even though all vaccines were tested in clinical trials prior to their general administration, serious adverse events, usually of very low incidence, were mostly identified after application of millions of doses. Establishing a direct correlation (the cause-effect paradigm) between vaccination and the appearance of adverse effects has proven challenging. This review focuses on the main adverse effects observed after vaccination, including anaphylaxis, myocarditis, vaccine-induced thrombotic thrombocytopenia, Guillain-Barré syndrome, and transverse myelitis reported in the context of COVID-19 vaccination. We highlight the symptoms, laboratory tests required for an adequate diagnosis, and briefly outline the recommended treatments for these adverse effects. The aim of this work is to increase awareness among healthcare personnel about the serious adverse events that may arise post-vaccination. Regardless of the ongoing discussion about the safety of COVID-19 vaccination, these adverse effects must be identified promptly and treated effectively to reduce the risk of complications.
Collapse
Affiliation(s)
- Teresa Padilla‐Flores
- Departamento de Biología Celular y del desarrollo, Instituto de Fisiología CelularUniversidad Nacional Autónoma de México (UNAM)Mexico CityMexico
| | - Alicia Sampieri
- Departamento de Biología Celular y del desarrollo, Instituto de Fisiología CelularUniversidad Nacional Autónoma de México (UNAM)Mexico CityMexico
| | - Luis Vaca
- Departamento de Biología Celular y del desarrollo, Instituto de Fisiología CelularUniversidad Nacional Autónoma de México (UNAM)Mexico CityMexico
| |
Collapse
|
12
|
Baykara N. Clinical Characteristics, Outcomes, and Risk Factors for Mortality in Pregnant/Puerperal Women with COVID-19 Admitted to ICU in Turkey: A Multicenter, Retrospective Study from a Middle-Income Country. J Intensive Care Med 2024; 39:577-594. [PMID: 38320979 DOI: 10.1177/08850666231222838] [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: 02/08/2024]
Abstract
BACKGROUND Coronaviruses have been the cause of 3 major outbreaks during the last 2 decades. Information on coronavirus diseases in pregnant women is limited, and even less is known about seriously ill pregnant women. Data are also lacking regarding the real burden of coronavirus disease 2019 (COVID-19) infection in pregnant women from low/middle-income countries. The aim of this study was to determine the characteristics and clinical course of COVID-19 in pregnant/puerperal women admitted to ICUs in Turkey. METHODS This was a national, multicenter, retrospective study. The study population comprised all SARS-CoV-2-infected pregnant/puerperal women admitted to participating ICUs between 1 March 2020 and 1 January 2022. Data regarding demographics, comorbidities, illness severity, therapies, extrapulmonary organ injuries, non-COVID-19 infections, and maternal and fetal/neonatal outcomes were recorded. LASSO logistic regression and multiple logistic regression analyses were used to identify predictive variables in terms of ICU mortality. RESULTS A total of 597 patients (341 pregnant women, 255 puerperal women) from 59 ICUs in 44 hospitals were included and of these patients, 87.1% were unvaccinated. The primary reason for ICU admission was acute hypoxemic respiratory failure in 522 (87.4%), acute hypoxemic respiratory failure plus shock in 14 (2.3%), ischemic cerebrovascular accident (CVA) in 5 (0.8%), preeclampsia/eclampsia/HELLP syndrome in 6 (1.0%), and post-caesarean follow-up in 36 (6.0%). Nonsurvivors were sicker than survivors upon ICU admission, with higher APACHE II (p < 0.001) and SOFA scores (p < 0.001). A total of 181 (30.3%) women died and 280 (46.6%) had received invasive mechanical ventilation (IMV). Myocardial injury, the highest SOFA score during ICU stay, LDH levels on admission, the highest levels of AST during ICU stay, average daily dose of corticosteroids, IMV, prophylactic dose anticoagulation (compared with therapeutic dose anticoagulation), PaO2/FiO2 ratio <100, pulmonary embolism, and shock were identified as predictors of mortality. Rates of premature birth (46.4%), cesarean section (53.7%), fetal distress (15.3%), stillbirth (6.5%), and low birth weight (19.4%) were high. Rates of neonatal death (8%) and respiratory distress syndrome (21%) were also high among live-born infants. CONCLUSIONS Severe/critical COVID-19 infection during the pregnancy/puerperal period was associated with high maternal mortality and fetal/neonatal complication rates in Turkey.
Collapse
Affiliation(s)
- Nur Baykara
- Department of Anesthesiology, Division of Critical Care, School of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
13
|
Verma VK, Yadav R, Beevi SS, Mohod AS, Mancharla S, Damodar N, Darapuneni RC, Reddy SG, Upendram P, Salt MN, Kulkarni P, Sahu S, Bollineni BR. Differential host responses to COVID-19: Unraveling the complexity. Diagn Microbiol Infect Dis 2024; 109:116281. [PMID: 38537507 DOI: 10.1016/j.diagmicrobio.2024.116281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/30/2024]
Abstract
These diverse outcomes of Covid-19 are influenced by various factors including age, gender, underlying health conditions, immune responses, viral variants, external factors, and overall quality of life. Demographic analysis of patients aged 0-18 years experienced mild to moderate cases, above 55 years with co-morbidities, were more severely affected.COVID-19 incidence was higher in males (58 %) & (42 %) in females. The reduced expression of Toll-like receptors (TLR) in severe and critical patients is a crucial determinant. This reduced TLR expression is primarily attributed to the dominance of the PLpro viral protein of COVID-19. Disease enrichment analysis highlights the long-term impact of COVID-19, which can lead to post-recovery complications such as hypertension, diabetes, cardiac diseases, and brain ischemia in Covid-19 patients. In conclusion, a comprehensive strategy targeting key factors like PLpro, TLR, and inflammatory cytokines such as IL-1 and IL-6 could offer an effective approach to mitigate the devastating effects of COVID-19.
Collapse
Affiliation(s)
- Vinod Kumar Verma
- KIMS Foundation and Research Centre, KIMS Hospital, Minister Road, Secunderabad, Telangana, India.
| | - Rahul Yadav
- KIMS Foundation and Research Centre, KIMS Hospital, Minister Road, Secunderabad, Telangana, India
| | - Syed Sultan Beevi
- KIMS Foundation and Research Centre, KIMS Hospital, Minister Road, Secunderabad, Telangana, India.
| | - Annie S Mohod
- KIMS Foundation and Research Centre, KIMS Hospital, Minister Road, Secunderabad, Telangana, India
| | - Shravya Mancharla
- KIMS Foundation and Research Centre, KIMS Hospital, Minister Road, Secunderabad, Telangana, India
| | - Neha Damodar
- KIMS Foundation and Research Centre, KIMS Hospital, Minister Road, Secunderabad, Telangana, India
| | | | - Sukurtha Gopal Reddy
- Diagnostics Division, Krishna Institute of Medical Sciences, Minister Road, Secunderabad, Telangana, India
| | - Pavani Upendram
- Department of Genetics, Krishna Institute of Medical Sciences, Minister Road, Secunderabad, Telangana, India
| | - Mohammad Nawaz Salt
- Department of Pulmonology, Krishna Institute of Medical Sciences, Minister Road, Secunderabad, Telangana, India
| | - Praveen Kulkarni
- Department of Internal Medicine, Krishna Institute of Medical Sciences, Minister Road, Secunderabad, Telangana, India
| | - Sambit Sahu
- Department of Critical care, Krishna Institute of Medical Sciences, Minister Road, Secunderabad, Telangana, India
| | - Bhaskar Rao Bollineni
- KIMS Foundation and Research Centre, KIMS Hospital, Minister Road, Secunderabad, Telangana, India
| |
Collapse
|
14
|
Li J, Huang Q, Liang Y, Jiang J, Yang Y, Feng J, Tan X, Li T. The Potential Mechanisms of Arrhythmia in Coronavirus disease-2019. Int J Med Sci 2024; 21:1366-1377. [PMID: 38818469 PMCID: PMC11134579 DOI: 10.7150/ijms.94578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) leads to coronavirus disease-2019 (COVID-19) which can cause severe cardiovascular complications including myocardial injury, arrhythmias, acute coronary syndrome and others. Among these complications, arrhythmias are considered serious and life-threatening. Although arrhythmias have been associated with factors such as direct virus invasion leading to myocardial injury, myocarditis, immune response disorder, cytokine storms, myocardial ischemia/hypoxia, electrolyte abnormalities, intravascular volume imbalances, drug interactions, side effects of COVID-19 vaccines and autonomic nervous system dysfunction, the exact mechanisms of arrhythmic complications in patients with COVID-19 are complex and not well understood. In the present review, the literature was extensively searched to investigate the potential mechanisms of arrhythmias in patients with COVID-19. The aim of the current review is to provide clinicians with a comprehensive foundation for the prevention and treatment of arrhythmias associated with long COVID-19.
Collapse
Affiliation(s)
- Jianhong Li
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qiuyuan Huang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Yifan Liang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Jian Feng
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
15
|
Al-Hassinah S, Al-Daihan S, Alahmadi M, Alghamdi S, Almulhim R, Obeid D, Arabi Y, Alswaji A, Aldriwesh M, Alghoribi M. Interplay of Demographic Influences, Clinical Manifestations, and Longitudinal Profile of Laboratory Parameters in the Progression of SARS-CoV-2 Infection: Insights from the Saudi Population. Microorganisms 2024; 12:1022. [PMID: 38792852 PMCID: PMC11124088 DOI: 10.3390/microorganisms12051022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Understanding the factors driving SARS-CoV-2 infection progression and severity is complex due to the dynamic nature of human physiology. Therefore, we aimed to explore the severity risk indicators of SARS-CoV-2 through demographic data, clinical manifestations, and the profile of laboratory parameters. The study included 175 patients either hospitalized at King Abdulaziz Medical City-Riyadh or placed in quarantine at designated hotels in Riyadh, Saudi Arabia, from June 2020 to April 2021. Hospitalized patients were followed up through the first week of admission. Demographic data, clinical presentations, and laboratory results were retrieved from electronic patient records. Our results revealed that older age (OR: 1.1, CI: [1.1-1.12]; p < 0.0001), male gender (OR: 2.26, CI: [1.0-5.1]; p = 0.047), and blood urea nitrogen level (OR: 2.56, CI: [1.07-6.12]; p = 0.034) were potential predictors of severity level. In conclusion, the study showed that apart from laboratory parameters, age and gender could potentially predict the severity of SARS-CoV-2 infection in the early stages. To our knowledge, this study is the first in Saudi Arabia to explore the longitudinal profile of laboratory parameters among risk factors, shedding light on SARS-CoV-2 infection progression parameters.
Collapse
Affiliation(s)
- Sarah Al-Hassinah
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Sooad Al-Daihan
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Mashael Alahmadi
- Research Office, Saudi National Institute of Health (SNIH), Riyadh 12382, Saudi Arabia;
| | - Sara Alghamdi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
| | - Rawabi Almulhim
- Infection Prevention and Control Department, King Abdulaziz Medical City, Riyadh 14611, Saudi Arabia;
| | - Dalia Obeid
- King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia;
| | - Yaseen Arabi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Intensive Care Department, King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Abdulrahman Alswaji
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
| | - Marwh Aldriwesh
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Majed Alghoribi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Department of Basic Science, College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| |
Collapse
|
16
|
Zhu Y, Cao X, Ying R, Liu K, Chai Y, Luo M, Huang Q, Gao P, Zhang C. Mapping the vast landscape of multisystem complications of COVID-19: Bibliometric analysis. Heliyon 2024; 10:e30760. [PMID: 38765136 PMCID: PMC11098853 DOI: 10.1016/j.heliyon.2024.e30760] [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: 11/01/2023] [Revised: 04/16/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Background With the rapid global spread of COVID-19, it has become evident that the virus can lead to multisystem complications, leading to a significant increase in related publications. Bibliometrics serves as a valuable tool for identifying highly cited literature and research hotspots within specific areas. Objective The aim of this study is to identify current research hotspots and future trends in COVID-19 complications. Methods The dataset was obtained from the Web of Science Core Collection, covering COVID-19 complications from December 8, 2019, to October 31, 2022. Various aspects, including publication general information, authors, journals, co-cited authors, co-cited references, research hotspots, and future trends, were subjected to analysis. Visual analysis was conducted using VOSviewer, The Online Analysis Platform of Literature Metrology, and Charticulator. Results There were 4597 articles in the study. The top three countries with the most published articles are the USA (n = 1350, 29.4 %), China (n = 765, 16.6 %), and Italy (n = 623, 13.6 %). USA and China have the closest collaborative relationship. The institute with the largest number of publications is Huazhong University of Science and Technology, followed by Harvard Medical School. Nevertheless, half of the top 10 institutes belong to the USA. "Rezaei, Nima" published 13 articles and ranked first, followed by "Yaghi, Shadi" with 12 articles and "Frontera, Jennifer" with 12 articles. The journal with the largest number of publications is "Journal of Clinical Medicine". The top 3 co-cited authors are "Zhou, Fei", "Guan, Wei-Jie", "Huang, Chaolin". The top 3 co-cited references addressed COVID-19's clinical features in China and noticed that COVID-19 patients had a wide range of complications. We also list four research hotspots. Conclusions This study conducted a bibliometric visual analysis of the literature on COVID-19 complications and summarized the current research hotspots. This study may provide valuable insights into the complications of COVID-19.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyu Cao
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rongtao Ying
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Liu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yilu Chai
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maocai Luo
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingsong Huang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peiyang Gao
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
17
|
Paes Leme AF, Yokoo S, Normando AGC, Ormonde JVS, Domingues RR, Cruz FF, Silva PL, Souza BSF, Dos Santos CC, Castro-Faria-Neto H, Martins CM, Lopes-Pacheco M, Rocco PRM. Proteomics of serum-derived extracellular vesicles are associated with the severity and different clinical profiles of patients with COVID-19: An exploratory secondary analysis. Cytotherapy 2024; 26:444-455. [PMID: 38363248 DOI: 10.1016/j.jcyt.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND AIMS Coronavirus disease 2019 (COVID-19) is characterized by a broad spectrum of clinical manifestations with the potential to progress to multiple organ dysfunction in severe cases. Extracellular vesicles (EVs) carry a range of biological cargoes, which may be used as biomarkers of disease state. METHODS An exploratory secondary analysis of the SARITA-2 and SARITA-1 datasets (randomized clinical trials on patients with mild and moderate/severe COVID-19) was performed. Serum-derived EVs were used for proteomic analysis to identify enriched biological processes and key proteins, thus providing insights into differences in disease severity. Serum-derived EVs were separated from patients with COVID-19 by size exclusion chromatography and nanoparticle tracking analysis was used to determine particle concentration and diameter. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was performed to identify and quantify protein signatures. Bioinformatics and multivariate statistical analysis were applied to distinguish candidate proteins associated with disease severity (mild versus moderate/severe COVID-19). RESULTS No differences were observed in terms of the concentration and diameter of enriched EVs between mild (n = 14) and moderate/severe (n = 30) COVID-19. A total of 414 proteins were found to be present in EVs, of which 360 were shared while 48 were uniquely present in severe/moderate compared to mild COVID-19. The main biological signatures in moderate/severe COVID-19 were associated with platelet degranulation, exocytosis, complement activation, immune effector activation, and humoral immune response. Von Willebrand factor, serum amyloid A-2 protein, histone H4 and H2A type 2-C, and fibrinogen β-chain were the most differentially expressed proteins between severity groups. CONCLUSION Exploratory proteomic analysis of serum-derived EVs from patients with COVID-19 detected key proteins related to immune response and activation of coagulation and complement pathways, which are associated with disease severity. Our data suggest that EV proteins may be relevant biomarkers of disease state and prognosis.
Collapse
Affiliation(s)
- Adriana F Paes Leme
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Sami Yokoo
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Ana Gabriela C Normando
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - João Vitor S Ormonde
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Romenia Ramos Domingues
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Fernanda F Cruz
- 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, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L Silva
- 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, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno S F Souza
- Goncalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil; D'Or Institute for Research and Education (IDOR), Salvador, Bahia, Brazil; Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Bahia, Brazil
| | - Claudia C Dos Santos
- The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Sciences and Interdepartmental Division of Critical Care, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Patricia R M 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, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
18
|
Cerneckis J, Cai H, Shi Y. Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications. Signal Transduct Target Ther 2024; 9:112. [PMID: 38670977 PMCID: PMC11053163 DOI: 10.1038/s41392-024-01809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
The induced pluripotent stem cell (iPSC) technology has transformed in vitro research and holds great promise to advance regenerative medicine. iPSCs have the capacity for an almost unlimited expansion, are amenable to genetic engineering, and can be differentiated into most somatic cell types. iPSCs have been widely applied to model human development and diseases, perform drug screening, and develop cell therapies. In this review, we outline key developments in the iPSC field and highlight the immense versatility of the iPSC technology for in vitro modeling and therapeutic applications. We begin by discussing the pivotal discoveries that revealed the potential of a somatic cell nucleus for reprogramming and led to successful generation of iPSCs. We consider the molecular mechanisms and dynamics of somatic cell reprogramming as well as the numerous methods available to induce pluripotency. Subsequently, we discuss various iPSC-based cellular models, from mono-cultures of a single cell type to complex three-dimensional organoids, and how these models can be applied to elucidate the mechanisms of human development and diseases. We use examples of neurological disorders, coronavirus disease 2019 (COVID-19), and cancer to highlight the diversity of disease-specific phenotypes that can be modeled using iPSC-derived cells. We also consider how iPSC-derived cellular models can be used in high-throughput drug screening and drug toxicity studies. Finally, we discuss the process of developing autologous and allogeneic iPSC-based cell therapies and their potential to alleviate human diseases.
Collapse
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Hongxia Cai
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
19
|
Hou Q, Jiang J, Na K, Zhang X, Liu D, Jing Q, Yan C, Han Y. Potential therapeutic targets for COVID-19 complicated with pulmonary hypertension: a bioinformatics and early validation study. Sci Rep 2024; 14:9294. [PMID: 38653779 DOI: 10.1038/s41598-024-60113-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
Coronavirus disease (COVID-19) and pulmonary hypertension (PH) are closely correlated. However, the mechanism is still poorly understood. In this article, we analyzed the molecular action network driving the emergence of this event. Two datasets (GSE113439 and GSE147507) from the GEO database were used for the identification of differentially expressed genes (DEGs).Common DEGs were selected by VennDiagram and their enrichment in biological pathways was analyzed. Candidate gene biomarkers were selected using three different machine-learning algorithms (SVM-RFE, LASSO, RF).The diagnostic efficacy of these foundational genes was validated using independent datasets. Eventually, we validated molecular docking and medication prediction. We found 62 common DEGs, including several ones that could be enriched for Immune Response and Inflammation. Two DEGs (SELE and CCL20) could be identified by machine-learning algorithms. They performed well in diagnostic tests on independent datasets. In particular, we observed an upregulation of functions associated with the adaptive immune response, the leukocyte-lymphocyte-driven immunological response, and the proinflammatory response. Moreover, by ssGSEA, natural killer T cells, activated dendritic cells, activated CD4 T cells, neutrophils, and plasmacytoid dendritic cells were correlated with COVID-19 and PH, with SELE and CCL20 showing the strongest correlation with dendritic cells. Potential therapeutic compounds like FENRETI-NIDE, AFLATOXIN B1 and 1-nitropyrene were predicted. Further molecular docking and molecular dynamics simulations showed that 1-nitropyrene had the most stable binding with SELE and CCL20.The findings indicated that SELE and CCL20 were identified as novel diagnostic biomarkers for COVID-19 complicated with PH, and the target of these two key genes, FENRETI-NIDE and 1-nitropyrene, was predicted to be a potential therapeutic target, thus providing new insights into the prediction and treatment of COVID-19 complicated with PH in clinical practice.
Collapse
Affiliation(s)
- Qingbin Hou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jinping Jiang
- Department of Cardiology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Kun Na
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaolin Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Quanmin Jing
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Yaling Han
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
20
|
Tang W, Ye B, Zhou L, Zou L. Risk prediction for severe COVID-19 progressing to critical illness and death in the ICU and efficacy analysis of using traditional Chinese medicine. Medicine (Baltimore) 2024; 103:e37498. [PMID: 38518027 PMCID: PMC10957017 DOI: 10.1097/md.0000000000037498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/14/2024] [Indexed: 03/24/2024] Open
Abstract
To reveal the key factors influencing the progression of severe COVID-19 to critical illness and death in the intensive care unit (ICU) and to accurately predict the risk, as well as to validate the efficacy of treatment using traditional Chinese medicine (TCM), thus providing valuable recommendations for the clinical management of patients. A total of 189 patients with COVID-19 in 25 ICUs in Chongqing, China, were enrolled, and 16 eventually died. Statistical models shown that factors influencing the progression of COVID-19 to critical illness include the severity of illness at diagnosis, the mode of respiratory support, and the use of TCM. Risk factors for death include a history of metabolic disease, the use of antiviral drugs and TCM, and invasive endotracheal intubation. The area under curve of the noncollinearity model predicted the risk of progression to critical illness and the risk of death reached 0.847 and 0.876, respectively. The use of TCM is an independent protective factor for the prevention of the progression of severe COVID-19, while uncorrectable hypoxemia and invasive respiratory support are independent risk factors, and antiviral drugs can help reduce mortality. The multifactorial prediction model can assess the risk of critical illness and death in ICU COVID-19 patients, and inform clinicians in choosing the treatment options and medications.
Collapse
Affiliation(s)
- Wenyi Tang
- Department of Clinical Data Research, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Bo Ye
- Department of Clinical Data Research, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Lina Zhou
- Department of Clinical Data Research, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Lingyun Zou
- Department of Clinical Data Research, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| |
Collapse
|
21
|
Trimarco V, Izzo R, Pacella D, Trama U, Manzi MV, Lombardi A, Piccinocchi R, Gallo P, Esposito G, Piccinocchi G, Lembo M, Morisco C, Rozza F, Santulli G, Trimarco B. Incidence of new-onset hypertension before, during, and after the COVID-19 pandemic: a 7-year longitudinal cohort study in a large population. BMC Med 2024; 22:127. [PMID: 38500180 PMCID: PMC10949764 DOI: 10.1186/s12916-024-03328-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND While the augmented incidence of diabetes after COVID-19 has been widely confirmed, controversial results are available on the risk of developing hypertension during the COVID-19 pandemic. METHODS We designed a longitudinal cohort study to analyze a closed cohort followed up over a 7-year period, i.e., 3 years before and 3 years during the COVID-19 pandemic, and during 2023, when the pandemic was declared to be over. We analyzed medical records of more than 200,000 adults obtained from a cooperative of primary physicians from January 1, 2017, to December 31, 2023. The main outcome was the new diagnosis of hypertension. RESULTS We evaluated 202,163 individuals in the pre-pandemic years and 190,743 in the pandemic years, totaling 206,857 when including 2023 data. The incidence rate of new hypertension was 2.11 (95% C.I. 2.08-2.15) per 100 person-years in the years 2017-2019, increasing to 5.20 (95% C.I. 5.14-5.26) in the period 2020-2022 (RR = 2.46), and to 6.76 (95% C.I. 6.64-6.88) in 2023. The marked difference in trends between the first and the two successive observation periods was substantiated by the fitted regression lines of two Poisson models conducted on the monthly log-incidence of hypertension. CONCLUSIONS We detected a significant increase in new-onset hypertension during the COVID-19 pandemic, which at the end of the observation period affected ~ 20% of the studied cohort, a percentage higher than the diagnosis of COVID-19 infection within the same time frame. This observation suggests that increased attention to hypertension screening should not be limited to individuals who are aware of having contracted the infection but should be extended to the entire population.
Collapse
Affiliation(s)
- Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences, and Dentistry, "Federico II" University, Naples, Italy
| | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Daniela Pacella
- Department of Public Health, "Federico II" University, Naples, Italy
| | - Ugo Trama
- Pharmaceutical Department of Campania Region, Naples, Italy
| | - Maria Virginia Manzi
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Angela Lombardi
- Department of Microbiology and Immunology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY, USA
| | | | - Paola Gallo
- Department of Neuroscience, Reproductive Sciences, and Dentistry, "Federico II" University, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Gaetano Piccinocchi
- COMEGEN Primary Care Physicians Cooperative, Italian Society of General Medicine (SIMG), Naples, Italy
| | - Maria Lembo
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Carmine Morisco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
- International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy
- Italian Society for Cardiovascular Prevention (SIPREC), Rome, Italy
| | - Francesco Rozza
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy.
- International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy.
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, 1300 Morris Park Avenue, New York City, NY, 10461, USA.
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
- International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy
- Italian Society for Cardiovascular Prevention (SIPREC), Rome, Italy
| |
Collapse
|
22
|
Del Vecchio L, Balafa O, Dounousi E, Ekart R, Fernandez BF, Mark PB, Sarafidis P, Valdivielso JM, Ferro CJ, Mallamaci F. COVID-19 and cardiovascular disease in patients with chronic kidney disease. Nephrol Dial Transplant 2024; 39:177-189. [PMID: 37771078 PMCID: PMC10828215 DOI: 10.1093/ndt/gfad170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Indexed: 09/30/2023] Open
Abstract
Millions of people worldwide have chronic kidney disease (CKD). Affected patients are at high risk for cardiovascular (CV) disease for several reasons. Among various comorbidities, CKD is associated with the more severe forms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This is particularly true for patients receiving dialysis or for kidney recipients. From the start of the SARS-CoV-2 pandemic, several CV complications have been observed in affected subjects, spanning acute inflammatory manifestations, CV events, thrombotic episodes and arrythmias. Several pathogenetic mechanisms have been hypothesized, including direct cytopathic viral effects on the myocardium, endothelial damage and hypercoagulability. This spectrum of disease can occur during the acute phase of the infection, but also months after recovery. This review is focussed on the CV complications of coronavirus disease 2019 (COVID-19) with particular interest in their implications for the CKD population.
Collapse
Affiliation(s)
- Lucia Del Vecchio
- Department of Nephrology and Dialysis, Sant'Anna Hospital, ASST Lariana, Como, Italy
| | - Olga Balafa
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | - Evangelia Dounousi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Medical Center Maribor, Maribor, Slovenia
| | | | - Patrick B Mark
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Pantelis Sarafidis
- 1st Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Jose M Valdivielso
- Vascular and Renal Translational Research Group, Institute for Biomedical Research on Lleida (IRBLleida), Lleida, Spain
| | - Charles J Ferro
- Department of Renal Medicine, University Hospitals Birmingham and Institute of Cardiovascular Sciences, University of Birmingham, Birmingham,UK
| | - Francesca Mallamaci
- Francesca Mallamaci Department of Nephrology, Dialysis, and Transplantation Azienda Ospedaliera “Bianchi-Melacrino-Morelli” & CNR-IFC, Reggio Calabria, Italy
| |
Collapse
|
23
|
Wang H, Wang Y. What Makes the Gut-Lung Axis Working? From the Perspective of Microbiota and Traditional Chinese Medicine. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:8640014. [PMID: 38274122 PMCID: PMC10810697 DOI: 10.1155/2024/8640014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
Background An increasing number of studies have proved that gut microbiota is involved in the occurrence and development of various lung diseases and can interact with the diseased lung. The concept of the gut-lung axis (GLA) provides a new idea for the subsequent clinical treatment of lung diseases through human microbiota. This review aims to summarize the microbiota in the lung and gut and the interaction between them from the perspectives of traditional Chinese medicine and modern medicine. Method We conducted a literature search by using the search terms "GLA," "gut microbiota," "spleen," and "Chinese medicine" in the databases PubMed, Web of Science, and CNKI. We then explored the mechanism of action of the gut-lung axis from traditional Chinese medicine and modern medicine. Results The lung and gut microbiota enable the GLA to function through immune regulation, while metabolites of the gut microbiota also play an important role. The spleen can improve the gut microbiota to achieve the regulation of the GLA. Conclusion Improving the gut microbiota through qi supplementation and spleen fortification provides a new approach to the clinical treatment of lung diseases by regulating the GLA. Currently, our understanding of the GLA is limited, and more research is needed to explain its working principle.
Collapse
Affiliation(s)
- Hui Wang
- Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Ying Wang
- Zhejiang Chinese Medical University, Hangzhou 310000, China
| |
Collapse
|
24
|
da Silva ALG, Vieira LDP, Dias LS, Prestes CV, Back GD, Goulart CDL, Arena R, Borghi-Silva A, Trimer R. Impact of long COVID on the heart rate variability at rest and during deep breathing maneuver. Sci Rep 2023; 13:22695. [PMID: 38123689 PMCID: PMC10733257 DOI: 10.1038/s41598-023-50276-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
While the majority of individuals with coronavirus disease 2019 (COVID-19) recover completely, a significant percentage experience persistent symptom, which has been characterized as Long COVID and may be associated with cardiac and autonomic dysfunction. We evaluated heart rate variability (HRV) at rest and during deep-breathing (M-RSA) in patients with Long COVID. Case-control design involved 21 patients with Long COVID and 20 controls; the HRV was evaluated (POLAR system) at rest in the supine position and during M-RSA and expressed in time domain and non-linear analysis. In the supine position we found a reduction HRV measures in Long COVID' patients compared to controls for: Mean_iRR (p < 0.001), STD_iRR (p < 0.001); STD_HR (p < 0.001); SD1 (p < 0.001); SD2 (p < 0.001); alpha2 (p < 0.001). In the M-RSA we found reduction Mean_iRR (p < 0.001), STD_iRR (p < 0.001), STD_HR (p < 0.001), rMSSD (p < 0.001), RR_tri-index (p < 0.001) in Long COVID' patients except for highest Mean_HR p < 0.001. In conclusion, Long COVID reduced HRV at rest and during deep breathing. These findings may imply impairment of cardiac autonomic control when symptoms of COVID-19 persist following initial recovery.
Collapse
Affiliation(s)
- Andréa Lúcia Gonçalves da Silva
- Department of Health Sciences, University of Santa Cruz do Sul, Rio Grande do Sul, Brazil.
- Nucleus of Research in Surveillance, Prevention and Rehabilitation in Cardiorespiratory Diseases, Rio Grande do Sul, Brazil.
| | - Luana Dos Passos Vieira
- Scientific Initiation Scholarship at Laboratory of Cardiorespiratory Rehabilitation, University of Santa Cruz do Sul, Rio Grande do Sul, Brazil
| | - Luiza Scheffer Dias
- Scientific Initiation Scholarship at Laboratory of Cardiorespiratory Rehabilitation, University of Santa Cruz do Sul, Rio Grande do Sul, Brazil
| | - Cecília Vieira Prestes
- Scientific Initiation Scholarship at Laboratory of Cardiorespiratory Rehabilitation, University of Santa Cruz do Sul, Rio Grande do Sul, Brazil
| | - Guilherme Dionir Back
- Cardiopulmonary Physiotherapy Laboratory, Physiotherapy Department, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | - Cassia da Luz Goulart
- Cardiopulmonary Physiotherapy Laboratory, Physiotherapy Department, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Audrey Borghi-Silva
- Cardiopulmonary Physiotherapy Laboratory, Physiotherapy Department, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | - Renata Trimer
- Department of Health Sciences, University of Santa Cruz do Sul, Rio Grande do Sul, Brazil
| |
Collapse
|
25
|
Sideratou CM, Papaneophytou C. Persisting Shadows: Unraveling the Impact of Long COVID-19 on Respiratory, Cardiovascular, and Nervous Systems. Infect Dis Rep 2023; 15:806-830. [PMID: 38131885 PMCID: PMC10742861 DOI: 10.3390/idr15060072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19), instigated by the zoonotic Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), rapidly transformed from an outbreak in Wuhan, China, into a widespread global pandemic. A significant post-infection condition, known as 'long- COVID-19' (or simply 'long- COVID'), emerges in a substantial subset of patients, manifesting with a constellation of over 200 reported symptoms that span multiple organ systems. This condition, also known as 'post-acute sequelae of SARS-CoV-2 infection' (PASC), presents a perplexing clinical picture with far-reaching implications, often persisting long after the acute phase. While initial research focused on the immediate pulmonary impact of the virus, the recognition of COVID-19 as a multiorgan disruptor has unveiled a gamut of protracted and severe health issues. This review summarizes the primary effects of long COVID on the respiratory, cardiovascular, and nervous systems. It also delves into the mechanisms underlying these impacts and underscores the critical need for a comprehensive understanding of long COVID's pathogenesis.
Collapse
Affiliation(s)
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus;
| |
Collapse
|
26
|
Zhang S, Liu X, Zhao Y, Wang P, Yu R, Xu P, Jiang Y, Cheng L. Microbiome characteristics description of COVID-19 patients based on bulk RNA-seq and scRNA-Seq data. Comput Biol Med 2023; 165:107400. [PMID: 37651767 DOI: 10.1016/j.compbiomed.2023.107400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/04/2023] [Accepted: 08/26/2023] [Indexed: 09/02/2023]
Abstract
After infection with SARS-CoV-2, the microbiome inside the human body changes dramatically. By re-annotating microbial sequences in bulk RNA-seq and scRNA-seq data of COVID-19 patients, we described the cellular microbial landscape of COVID-19 patients and identified characteristic microorganisms in various tissues. We found that Acinetobacter lwoffii was highly correlated with COVID-19 symptoms and might disrupt some pathways of patients by interacting with the host and other microbes, such as Klebsiella pneumoniae. We further identified characteristic microorganisms specific to cell type, indicating the enrichment preference of some microbes. We also revealed the co-infection of SARS-CoV-2 with hMPV, which may cause the development of COVID-19. Overall, we demonstrated that the presence of intracellular microorganisms in COVID-19 patients and the synergies between microorganisms were strongly correlated with disease progression, providing a theoretical basis for COVID-19 treatment in a certain extent.
Collapse
Affiliation(s)
- Sainan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| | - Xingwang Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| | - Yue Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| | - Ping Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| | - Rui Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| | - Peigang Xu
- Chongqing Research Institute of Harbin Institute of Technology, China.
| | - Yue Jiang
- Cipher Gene, Ltd., Beijing, 100080, China.
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China; NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150028, Heilongjiang, China.
| |
Collapse
|
27
|
Milivojević V, Bogdanović J, Babić I, Todorović N, Ranković I. Metabolic Associated Fatty Liver Disease (MAFLD) and COVID-19 Infection: An Independent Predictor of Poor Disease Outcome? MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1438. [PMID: 37629728 PMCID: PMC10456234 DOI: 10.3390/medicina59081438] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023]
Abstract
Background and Objectives: Early reports on COVID-19 infection suggested that the SARS-CoV-2 virus solely attacks respiratory tract cells. As the pandemic spread, it became clear that the infection is multiorganic. Metabolic associated fatty liver disease (MAFLD) is a chronic liver disease strongly associated with insulin resistance and diabetes. The aim of this study was to assess a possible interplay between MAFLD and COVID-19 infection and its implication in COVID-19 outcome. Materials and Methods: A retrospective observational study, including 130 COVID-19 positive patients was conducted. MAFLD diagnosis was made based on the International Consensus criteria. Patients were divided into two groups, group A (MAFLD) and group B (nonMAFLD). Anthropometric and laboratory analysis were obtained. COVID-19 severity was assessed using the NEWS2 score. Disease outcome was threefold and regarded as discharged, patients who required mechanical ventilation (MV), and deceased patients. Results: MAFLD prevalence was 42%, 67% of patients were discharged, and 19% needed MV. Mortality rate was 14%. MAFLD patients were significantly younger (p < 0.001), and had higher body mass index (p < 0.05), respiratory rate (p < 0.05) and systolic blood pressure (p < 0.05) than nonMAFLD patients. Regarding metabolic syndrome and inflammatory markers: group A had significantly higher glycemia at admission (p = 0.008), lower HDL-c (p < 0.01), higher triglycerides (p < 0.01), CRP (p < 0.001), IL-6 (p < 0.05) and ferritin (p < 0.05) than group B. MAFLD was associated with more prevalent type 2 diabetes (p = 0.035) and hypertension (p < 0.05). MAFLD patients had a more severe disease course (NEWS2 score, 6.5 ± 0.5 vs. 3 ± 1.0, p < 0.05). MAFLD presence was associated with lower patient discharge (p < 0.01) and increased need for MV (p = 0.024). Multiple regression analysis showed that BMI (p = 0.045), IL-6 (p = 0.03), and MAFLD (p < 0.05) are significant independent risk factors for a poor COVID-19 outcome. Conclusions: The prevalence of MAFLD is relatively high. MAFLD patients had a more severe COVID-19 clinical course and worse disease outcome. Our results imply that early patient stratification and risk assessment are mandatory in order to avoid poor outcomes.
Collapse
Affiliation(s)
- Vladimir Milivojević
- Clinic for Gastroenterology and Hepatology University Clinical Centre of Serbia, Dr Koste Todorovica 2, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia
| | - Jelena Bogdanović
- Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia
- Clinic for Endocrinology, Diabetes and Metabolic Diseases University Clinical Centre of Serbia, Dr Subotica 13, 11000 Belgrade, Serbia
| | - Ivana Babić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases University Clinical Centre of Serbia, Dr Subotica 13, 11000 Belgrade, Serbia
| | - Nevena Todorović
- Clinic for Infectious and Tropical Diseases University Clinical Centre of Serbia, Bulevar Oslobođenja 16, 11000 Belgrade, Serbia
| | - Ivan Ranković
- Department of Gastroenterology, Royal Cornwall Hospitals NHS Trust, Truro TR1 3LJ, UK;
| |
Collapse
|
28
|
Fedeli U, Barbiellini Amidei C, Casotto V, Grande E, Saia M, Zanetto A, Russo FP. Mortality from chronic liver disease: Recent trends and impact of the COVID-19 pandemic. World J Gastroenterol 2023; 29:4166-4173. [PMID: 37475843 PMCID: PMC10354574 DOI: 10.3748/wjg.v29.i26.4166] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/26/2023] [Accepted: 06/12/2023] [Indexed: 07/10/2023] Open
Abstract
Prepandemic time trends in mortality from chronic liver disease (CLD) differed according to specific cause of death (decreasing for liver cirrhosis, stable or increasing for liver cancer), etiology (increasing for nonalcoholic fatty liver disease, generally decreasing for other etiologies), and world region (decreasing in areas with the highest burden of hepatitis B virus, increasing in Eastern Europe and other countries). The coronavirus disease 2019 (COVID-19) pandemic affected mortality of patients with CLD both directly, with a higher risk for severe illness and death depending on age, stage and etiology of the disease, and indirectly, through social isolation and loss of support, harmful drinking, and difficulties in access to care. Nevertheless, only sparse data are available on variations in CLD as a cause of death during the pandemic. In the USA, in 2020-2021 a growth in mortality was registered for all liver diseases, more marked for alcoholic liver disease, especially among young people aged 25-44 years and in selected ethnic groups. COVID-19 related deaths accounted only for a minor part of the excess. Further data from mortality registers of other countries are warranted, preferably adopting the so-called multiple cause-of-death approach, and extended to deaths attributed to viral hepatitis and liver cancer.
Collapse
Affiliation(s)
- Ugo Fedeli
- Epidemiological Department, Azienda Zero, Veneto Region, Padova 35132, Italy
| | | | - Veronica Casotto
- Epidemiological Department, Azienda Zero, Veneto Region, Padova 35132, Italy
| | - Enrico Grande
- Integrated System for Health, Social Assistance and Welfare, National Institute of Statistics, Rome 00184, Italy
| | - Mario Saia
- Clinical Governance, Azienda Zero, Veneto Region, Padova 35132, Italy
| | - Alberto Zanetto
- Department of Surgery Oncology and Gastroenterology, University of Padua, Padova 35128, Italy
- Gastroenterology and Multivisceral Transplant Unit, Azienda Ospedale-Università di Padova, Padova 25128, Italy
| | - Francesco Paolo Russo
- Department of Surgery Oncology and Gastroenterology, University of Padua, Padova 35128, Italy
- Gastroenterology and Multivisceral Transplant Unit, Azienda Ospedale-Università di Padova, Padova 25128, Italy
| |
Collapse
|
29
|
König S, Vollenberg R, Tepasse PR. The Renin-Angiotensin System in COVID-19: Can Long COVID Be Predicted? Life (Basel) 2023; 13:1462. [PMID: 37511837 PMCID: PMC10381802 DOI: 10.3390/life13071462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: Co-morbidities such as hypertension and cardiovascular disease are major risk factors for severe COVID-19. The renin-angiotensin system (RAS) is critically involved in their pathophysiology and is counter-balanced by both angiotensin-converting enzyme 2 (ACE2), the functional receptor of SARS-CoV-2, and the kallikrein-kinin system (KKS). Considerable research interest with respect to COVID-19 treatment is currently being directed towards the components of these systems. In earlier studies, we noticed significantly reduced carboxypeptidase N (CPN, KKS member) activity and excessive angiotensin-converting enzyme (ACE, RAS member) activity in the sera of both hospitalized COVID-19 patients and a subgroup of convalescent patients. The data had been obtained using labeled bradykinin (BK) as a reporter peptide, which is a target of both CPN and ACE. The data were supplemented with mass-spectrometry-based serum proteomic analysis. Here, we hypothesize that the degree of BK serum degradation could be indicative of Long COVID. (2) Review and Discussion: The recent literature is briefly reviewed. The fact that the levels of the BK serum degradation products did not reach normal concentrations in almost half of the patients during convalescences could have been partially due to a dysregulated RAS. (3) Conclusions: Standard tests for routine patient care in Long COVID come often back normal. We suggest that the measurement of selected members of the RAS such as ACE and angiotensin II or the use of our BK degradation assay could identify Long COVID candidates. Clinical studies are required to test this hypothesis.
Collapse
Affiliation(s)
- Simone König
- IZKF Core Unit Proteomics, University of Münster, 48149 Münster, Germany
| | - Richard Vollenberg
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Muenster, 48149 Münster, Germany
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Muenster, 48149 Münster, Germany
| |
Collapse
|
30
|
Pieri M, Vayianos P, Nicolaidou V, Felekkis K, Papaneophytou C. Alterations in Circulating miRNA Levels after Infection with SARS-CoV-2 Could Contribute to the Development of Cardiovascular Diseases: What We Know So Far. Int J Mol Sci 2023; 24:ijms24032380. [PMID: 36768701 PMCID: PMC9917196 DOI: 10.3390/ijms24032380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/09/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and poses significant complications for cardiovascular disease (CVD) patients. MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression and influence several physiological and pathological processes, including CVD. This critical review aims to expand upon the current literature concerning miRNA deregulation during the SARS-CoV-2 infection, focusing on cardio-specific miRNAs and their association with various CVDs, including cardiac remodeling, arrhythmias, and atherosclerosis after SARS-CoV-2 infection. Despite the scarcity of research in this area, our findings suggest that changes in the expression levels of particular COVID-19-related miRNAs, including miR-146a, miR-27/miR-27a-5p, miR-451, miR-486-5p, miR-21, miR-155, and miR-133a, may be linked to CVDs. While our analysis did not conclusively determine the impact of SARS-CoV-2 infection on the profile and/or expression levels of cardiac-specific miRNAs, we proposed a potential mechanism by which the miRNAs mentioned above may contribute to the development of these two pathologies. Further research on the relationship between SARS-CoV-2, CVDs, and microRNAs will significantly enhance our understanding of this connection and may lead to the use of these miRNAs as biomarkers or therapeutic targets for both pathologies.
Collapse
Affiliation(s)
- Myrtani Pieri
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
- Non-Coding RNA Research Laboratory, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
| | - Panayiotis Vayianos
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
| | - Vicky Nicolaidou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
- Non-Coding RNA Research Laboratory, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
| | - Kyriacos Felekkis
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
- Non-Coding RNA Research Laboratory, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
- Correspondence: (K.F.); (C.P.)
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
- Non-Coding RNA Research Laboratory, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
- Correspondence: (K.F.); (C.P.)
| |
Collapse
|
31
|
Motallebnezhad M, Omraninava M, Esmaeili Gouvarchin Ghaleh H, Jonaidi-Jafari N, Hazrati A, Malekpour K, Bagheri Y, Izadi M, Ahmadi M. Potential therapeutic applications of extracellular vesicles in the immunopathogenesis of COVID-19. Pathol Res Pract 2023; 241:154280. [PMID: 36580795 PMCID: PMC9759301 DOI: 10.1016/j.prp.2022.154280] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the cause of coronavirus disease 2019 (COVID-19) which has emerged as a global health crisis. Recently, more than 50 different types of potential COVID-19 vaccines have been developed to elicit a strong immune response against SARS-CoV-2. However, genetic mutations give rise to the new variants of SARS-CoV-2 which is highly associated with the reduced effectiveness of COVID-19 vaccines. There is still no efficient antiviral agent to specifically target the SARS-CoV-2 infection and treatment of COVID-19. Therefore, understanding the molecular mechanisms underlying the pathogenesis of SARS-CoV-2 may contribute to discovering a novel potential therapeutic approach to the management of COVID-19. Recently, extracellular vesicle (EV)-based therapeutic strategies have received great attention on account of their potential benefits in the administration of viral diseases. EVs are extracellular vesicles containing specific biomolecules which play an important role in cell-to-cell communications. It has been revealed that EVs are involved in the pathogenesis of different inflammatory diseases such as cancer and viral infections. EVs are released from virus-infected cells which could mediate the interaction of infected and uninfected host cells. Hence, these extracellular nanoparticles have been considered a novel approach for drug delivery to mediate the treatment of a wide range of diseases including, COVID-19. EVs are considered a cell-free therapeutic strategy that could ameliorate the cytokine storm and its complications in COVID-19 patients. Furthermore, EV-based cargo delivery such as immunomodulatory agents in combination with antiviral drugs may have therapeutic benefits in patients with SARS-CoV-2 infection. In this review, we will highlight the potential of EVs as a therapeutic candidate in the diagnosis and treatment of COVID-19. Also, we will discuss the future perspectives regarding the beneficial effects of Evs in the development of COVID-19 vaccines.
Collapse
Affiliation(s)
- Morteza Motallebnezhad
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Melodi Omraninava
- Department of Infectious Disease, Faculty of Medical Sciences, Sari Branch, Islamic Azad University, Sari, Iran
| | | | - Nematollah Jonaidi-Jafari
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Yasser Bagheri
- Immunology Department, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Morteza Izadi
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
32
|
Arish M, Qian W, Narasimhan H, Sun J. COVID-19 immunopathology: From acute diseases to chronic sequelae. J Med Virol 2023; 95:e28122. [PMID: 36056655 PMCID: PMC9537925 DOI: 10.1002/jmv.28122] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 01/17/2023]
Abstract
The clinical manifestation of coronavirus disease 2019 (COVID-19) mainly targets the lung as a primary affected organ, which is also a critical site of immune cell activation by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, recent reports also suggest the involvement of extrapulmonary tissues in COVID-19 pathology. The interplay of both innate and adaptive immune responses is key to COVID-19 management. As a result, a robust innate immune response provides the first line of defense, concomitantly, adaptive immunity neutralizes the infection and builds memory for long-term protection. However, dysregulated immunity, both innate and adaptive, can skew towards immunopathology both in acute and chronic cases. Here we have summarized some of the recent findings that provide critical insight into the immunopathology caused by SARS-CoV-2, in acute and post-acute cases. Finally, we further discuss some of the immunomodulatory drugs in preclinical and clinical trials for dampening the immunopathology caused by COVID-19.
Collapse
Affiliation(s)
- Mohd Arish
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Wei Qian
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Harish Narasimhan
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Jie Sun
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
33
|
Khondakar KR, Kaushik A. Role of Wearable Sensing Technology to Manage Long COVID. BIOSENSORS 2022; 13:62. [PMID: 36671900 PMCID: PMC9855989 DOI: 10.3390/bios13010062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 06/17/2023]
Abstract
Long COVID consequences have changed the perception towards disease management, and it is moving towards personal healthcare monitoring. In this regard, wearable devices have revolutionized the personal healthcare sector to track and monitor physiological parameters of the human body continuously. This would be largely beneficial for early detection (asymptomatic and pre-symptomatic cases of COVID-19), live patient conditions, and long COVID monitoring (COVID recovered patients and healthy individuals) for better COVID-19 management. There are multitude of wearable devices that can observe various human body parameters for remotely monitoring patients and self-monitoring mode for individuals. Smart watches, smart tattoos, rings, smart facemasks, nano-patches, etc., have emerged as the monitoring devices for key physiological parameters, such as body temperature, respiration rate, heart rate, oxygen level, etc. This review includes long COVID challenges for frequent monitoring of biometrics and its possible solution with wearable device technologies for diagnosis and post-therapy of diseases.
Collapse
Affiliation(s)
- Kamil Reza Khondakar
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun 248007, Uttarakhand, India
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805-8531, USA
- Department of Chemical Engineering, University of Johannesburg, Johannesburg 2094, South Africa
| |
Collapse
|
34
|
Xian J, Huang H, Huang G, Zhou R, Yang M, Qiu Y, Bi L, Su Z, Xiao F, Shan H, Jin H. A Positron Emission Tomography Tracer Targeting the S2 Subunit of SARS-CoV-2 in Extrapulmonary Infections. Mol Pharm 2022; 19:4264-4274. [PMID: 36067000 PMCID: PMC9469952 DOI: 10.1021/acs.molpharmaceut.2c00584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/28/2022]
Abstract
Tracking the pathogen of coronavirus disease 2019 (COVID-19) in live subjects may help estimate the spatiotemporal distribution of SARS-CoV-2 infection in vivo. This study developed a positron emission tomography (PET) tracer of the S2 subunit of spike (S) protein for imaging SARS-CoV-2. A pan-coronavirus inhibitor, EK1 peptide, was synthesized and radiolabeled with copper-64 after being conjugated with 1,4,7-triazacyclononane-1,4,7-triyl-triacetic acid (NOTA). The in vitro stability tests indicated that [64Cu]Cu-NOTA-EK1 was stable up to 24 h both in saline and in human serum. The binding assay showed that [64Cu]Cu-NOTA-EK1 has a nanomolar affinity (Ki = 3.94 ± 0.51 nM) with the S-protein of SARS-CoV-2. The cell uptake evaluation used HEK293T/S+ and HEK293T/S- cell lines that showed that the tracer has a high affinity with the S-protein on the cellular level. For the in vivo study, we tested [64Cu]Cu-NOTA-EK1 in HEK293T/S+ cell xenograft-bearing mice (n = 3) and pseudovirus of SARS-CoV-2-infected HEK293T/ACE2 cell bearing mice (n = 3). The best radioactive xenograft-to-muscle ratio (X/Nxenograft 8.04 ± 0.99, X/Npseudovirus 6.47 ± 0.71) was most evident 4 h postinjection. Finally, PET imaging in the surrogate mouse model of beta-coronavirus, mouse hepatic virus-A59 infection in C57BL/6 J mice showed significantly enhanced accumulation in the liver than in the uninfected mice (1.626 ± 0.136 vs 0.871 ± 0.086 %ID/g, n = 3, P < 0.05) at 4 h postinjection. In conclusion, our experimental results demonstrate that [64Cu]Cu-NOTA-EK1 is a potential molecular imaging probe for tracking SARS-CoV-2 in extrapulmonary infections in living subjects.
Collapse
Affiliation(s)
- Jianzhong Xian
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
- Department of Ultrasound Medicine, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong
Province519000, China
| | - Hongbin Huang
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
| | - Guolong Huang
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
| | - Renwei Zhou
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
| | - Min Yang
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
| | - Yifan Qiu
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
| | - Lei Bi
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
| | - Zhongzhen Su
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
- Department of Ultrasound Medicine, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong
Province519000, China
| | - Fei Xiao
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
| | - Hong Shan
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
- Department of Interventional Medicine, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong
Province519000, China
| | - Hongjun Jin
- Guangdong Provincial Key Laboratory of Biomedical
Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University,
Zhuhai, Guangdong Province519000, China
| |
Collapse
|
35
|
Pelle MC, Zaffina I, Lucà S, Forte V, Trapanese V, Melina M, Giofrè F, Arturi F. Endothelial Dysfunction in COVID-19: Potential Mechanisms and Possible Therapeutic Options. Life (Basel) 2022; 12:1605. [PMID: 36295042 PMCID: PMC9604693 DOI: 10.3390/life12101605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/29/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
SARS-CoV-2, a novel coronavirus found in Wuhan (China) at the end of 2019, is the etiological agent of the current pandemic that is a heterogeneous disease, named coronavirus disease 2019 (COVID-19). SARS-CoV-2 affects primarily the lungs, but it can induce multi-organ involvement such as acute myocardial injury, myocarditis, thromboembolic eventsandrenal failure. Hypertension, chronic kidney disease, diabetes mellitus and obesity increase the risk of severe complications of COVID-19. There is no certain explanation for this systemic COVID-19 involvement, but it could be related to endothelial dysfunction, due to direct (endothelial cells are infected by the virus) and indirect damage (systemic inflammation) factors. Angiotensin-converting enzyme 2 (ACE2), expressed in human endothelium, has a fundamental role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In fact, ACE2 is used as a receptor by SARS-CoV-2, leading to the downregulation of these receptors on endothelial cells; once inside, this virus reduces the integrity of endothelial tissue, with exposure of prothrombotic molecules, platelet adhesion, activation of coagulation cascades and, consequently, vascular damage. Systemic microangiopathy and thromboembolism can lead to multi-organ failure with an elevated risk of death. Considering the crucial role of the immunological response and endothelial damage in developing the severe form of COVID-19, in this review, we will attempt to clarify the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Maria Chiara Pelle
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Isabella Zaffina
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Stefania Lucà
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Valentina Forte
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Trapanese
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Melania Melina
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Federica Giofrè
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Franco Arturi
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Research Centre for the Prevention and Treatment of Metabolic Diseases (CR METDIS), University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
36
|
Forsyth CB, Zhang L, Bhushan A, Swanson B, Zhang L, Mamede JI, Voigt RM, Shaikh M, Engen PA, Keshavarzian A. The SARS-CoV-2 S1 Spike Protein Promotes MAPK and NF-kB Activation in Human Lung Cells and Inflammatory Cytokine Production in Human Lung and Intestinal Epithelial Cells. Microorganisms 2022; 10:microorganisms10101996. [PMID: 36296272 PMCID: PMC9607240 DOI: 10.3390/microorganisms10101996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/12/2022] [Accepted: 10/05/2022] [Indexed: 11/18/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic began in January 2020 in Wuhan, China, with a new coronavirus designated SARS-CoV-2. The principal cause of death from COVID-19 disease quickly emerged as acute respiratory distress syndrome (ARDS). A key ARDS pathogenic mechanism is the “Cytokine Storm”, which is a dramatic increase in inflammatory cytokines in the blood. In the last two years of the pandemic, a new pathology has emerged in some COVID-19 survivors, in which a variety of long-term symptoms occur, a condition called post-acute sequelae of COVID-19 (PASC) or “Long COVID”. Therefore, there is an urgent need to better understand the mechanisms of the virus. The spike protein on the surface of the virus is composed of joined S1–S2 subunits. Upon S1 binding to the ACE2 receptor on human cells, the S1 subunit is cleaved and the S2 subunit mediates the entry of the virus. The S1 protein is then released into the blood, which might be one of the pivotal triggers for the initiation and/or perpetuation of the cytokine storm. In this study, we tested the hypothesis that the S1 spike protein is sufficient to activate inflammatory signaling and cytokine production, independent of the virus. Our data support a possible role for the S1 spike protein in the activation of inflammatory signaling and cytokine production in human lung and intestinal epithelial cells in culture. These data support a potential role for the SARS-CoV-2 S1 spike protein in COVID-19 pathogenesis and PASC.
Collapse
Affiliation(s)
- Christopher B. Forsyth
- Department of Internal Medicine, Section of Gastroenterology, Rush Center for Integrated Microbiome and Chronobiology Research, Department of Anatomy and Cell Biology, Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612, USA
| | - Lijuan Zhang
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA
| | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Barbara Swanson
- Department of Adult Health & Gerontological Nursing, Rush University Medical Center, Chicago, IL 60612, USA
| | - Li Zhang
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - João I. Mamede
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Robin M. Voigt
- Department of Internal Medicine, Section of Gastroenterology, Rush Center for Integrated Microbiome and Chronobiology Research, Department of Anatomy and Cell Biology, Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612, USA
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA
| | - Phillip A. Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ali Keshavarzian
- Department of Internal Medicine, Section of Gastroenterology, Rush Center for Integrated Microbiome and Chronobiology Research, Department of Anatomy and Cell Biology, Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612, USA
- Correspondence:
| |
Collapse
|
37
|
Tufa A, Gebremariam TH, Manyazewal T, Asrat Y, Getinet T, Hundie TG, Webb DL, Hellström PM, Genet S. Limited value of neutrophil-to-lymphocyte ratio and serum creatinine as point-of-care biomarkers of disease severity and infection mortality in patients hospitalized with COVID-19. PLoS One 2022; 17:e0275391. [PMID: 36201435 PMCID: PMC9536552 DOI: 10.1371/journal.pone.0275391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/15/2022] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION In hospitalized COVID-19, neutrophil-to-lymphocyte ratio (NLR) and serum creatinine is sometimes measured under assumption they predict disease severity and mortality. We determined the potential value of NLR and serum creatinine as predictors of disease severity and mortality in COVID-19. METHODS Prospective cohort study of COVID-19 patients admitted to premier COVID-19 treatment hospitals in Ethiopia. Predictive capability of biomarkers in progression and prognosis of COVID-19 was analyzed using receiver operating characteristics. Survival of COVID-19 patients with different biomarker levels was computed. Logistic regression assessed associations between disease severity and mortality on NLR and serum creatinine adjusted for odds ratio (AOR). RESULTS The study enrolled 126 adults with severe (n = 68) or mild/moderate (n = 58) COVID-19, with median age 50 [interquartile range (IQR 20-86)]; 57.1% males. The NLR value was significantly higher in severe cases [6.68 (IQR 3.03-12.21)] compared to the mild/moderate [3.23 (IQR 2.09-5.39)], with the NLR value markedly associated with disease severity (p<0.001). Mortality was higher in severe cases [13 (19.1%)] compared to mild/moderate cases [2 (3.4%)] (p = 0.007). The NLR value was significantly higher in non-survivors [15.17 (IQR 5.13-22.5)] compared to survivors [4.26 (IQR 2.40-7.90)] (p = 0.002). Serum creatinine was significantly elevated in severe cases [34 (50%)] compared with mild/moderate [11 (19%)] (p<0.001). Disease severity [AOR 6.58, 95%CI (1.29-33.56), p = 0.023] and NLR [AOR 1.07, 95%CI (1.02-1.12), p = 0.004)] might be associated with death. NLR had a sensitivity and specificity of 69.1% and 60.3% as predictor of disease severity (cut-off >4.08), and 86.7% and 55.9% as prognostic marker of mortality (cut-off >4.63). CONCLUSION In COVID-19, NLR is a biomarker with only modest accuracy for predicting disease severity and mortality. Still, patients with NLR >4.63 are more likely to die. Monitoring of this biomarker at the earliest stage of the disease may predict outcome. Additionally, high creatinine seems related to disease severity and mortality.
Collapse
Affiliation(s)
- Abdisa Tufa
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tewodros Haile Gebremariam
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tsegahun Manyazewal
- Centre for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Yidnekachew Asrat
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tewodros Getinet
- School of Public Health, Saint Paul’s Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | | | - Dominic-Luc Webb
- Gastroenterology and Hepatology Unit, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Per M. Hellström
- Gastroenterology and Hepatology Unit, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Solomon Genet
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
38
|
da Silva SJR, do Nascimento JCF, Germano Mendes RP, Guarines KM, Targino Alves da Silva C, da Silva PG, de Magalhães JJF, Vigar JRJ, Silva-Júnior A, Kohl A, Pardee K, Pena L. Two Years into the COVID-19 Pandemic: Lessons Learned. ACS Infect Dis 2022; 8:1758-1814. [PMID: 35940589 PMCID: PMC9380879 DOI: 10.1021/acsinfecdis.2c00204] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly transmissible and virulent human-infecting coronavirus that emerged in late December 2019 in Wuhan, China, causing a respiratory disease called coronavirus disease 2019 (COVID-19), which has massively impacted global public health and caused widespread disruption to daily life. The crisis caused by COVID-19 has mobilized scientists and public health authorities across the world to rapidly improve our knowledge about this devastating disease, shedding light on its management and control, and spawned the development of new countermeasures. Here we provide an overview of the state of the art of knowledge gained in the last 2 years about the virus and COVID-19, including its origin and natural reservoir hosts, viral etiology, epidemiology, modes of transmission, clinical manifestations, pathophysiology, diagnosis, treatment, prevention, emerging variants, and vaccines, highlighting important differences from previously known highly pathogenic coronaviruses. We also discuss selected key discoveries from each topic and underline the gaps of knowledge for future investigations.
Collapse
Affiliation(s)
- Severino Jefferson Ribeiro da Silva
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Pernambuco, Brazil.,Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Jessica Catarine Frutuoso do Nascimento
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Pernambuco, Brazil
| | - Renata Pessôa Germano Mendes
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Pernambuco, Brazil
| | - Klarissa Miranda Guarines
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Pernambuco, Brazil
| | - Caroline Targino Alves da Silva
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Pernambuco, Brazil
| | - Poliana Gomes da Silva
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Pernambuco, Brazil
| | - Jurandy Júnior Ferraz de Magalhães
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Pernambuco, Brazil.,Department of Virology, Pernambuco State Central Laboratory (LACEN/PE), 52171-011 Recife, Pernambuco, Brazil.,University of Pernambuco (UPE), Serra Talhada Campus, 56909-335 Serra Talhada, Pernambuco, Brazil.,Public Health Laboratory of the XI Regional Health, 56912-160 Serra Talhada, Pernambuco, Brazil
| | - Justin R J Vigar
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Abelardo Silva-Júnior
- Institute of Biological and Health Sciences, Federal University of Alagoas (UFAL), 57072-900 Maceió, Alagoas, Brazil
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, United Kingdom
| | - Keith Pardee
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada.,Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Lindomar Pena
- Laboratory of Virology and Experimental Therapy (LAVITE), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Pernambuco, Brazil
| |
Collapse
|
39
|
Cui H, Wang Y, Yu B, Wu Y, Zhang G, Guo J, Luo J, Li Q, Li X, He W, Wen W, Liao J, Wang D. Jian-Ti-Kang-Yi decoction alleviates poly(I:C)-induced pneumonia by inhibiting inflammatory response, reducing oxidative stress, and modulating host metabolism. Front Pharmacol 2022; 13:979400. [PMID: 36147321 PMCID: PMC9486163 DOI: 10.3389/fphar.2022.979400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Jian-Ti-Kang-Yi decoction (JTKY) is widely used in the treatment of COVID-19. However, the protective mechanisms of JTKY against pneumonia remain unknown. In this study, polyinosinic-polycytidylic acid (poly(I:C)), a mimic of viral dsRNA, was used to induce pneumonia in mice; the therapeutic effects of JTKY on poly(I:C)-induced pneumonia model mice were evaluated. In addition, the anti-inflammatory and anti-oxidative potentials of JTKY were also investigated. Lastly, the metabolic regulatory effects of JTKY in poly(I:C)-induced pneumonia model mice were studied using untargeted metabolomics. Our results showed that JTKY treatment decreased the wet-to-dry ratio in the lung tissue, total protein concentration, and total cell count of the bronchoalveolar lavage fluid (BALF). Hematoxylin and Eosin (HE) and Masson staining indicated that the JTKY treatment alleviated the pathological changes and decreased the fibrotic contents in the lungs. JTKY treatment also decreased the expression of pro-inflammatory cytokines [interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α)] and increased the levels of immunomodulatory cytokines (IL-4 and IL-10) in the BALF and serum. Flow cytometry analysis showed that the JTKY treatment lowered the ratio of CD86+/CD206+ macrophages in the BALF, decreased inducible nitric oxide synthase (iNOS) level, and increased arginase 1 (Arg-1) level in lung. JTKY also lowered CD11b+Ly6G+ neutrophils in BALF and decreased myeloperoxidase (MPO) activity in lung. Moreover, it also elevated superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities and decreased methane dicarboxylic aldehyde (MDA) level in lung. Untargeted metabolomic analysis showed that the JTKY treatment could affect 19 metabolites in lung, such as L-adrenaline, L-asparagine, ornithine, and alpha-ketoglutaric acid. These metabolites are associated with the synthesis and degradation of ketone bodies, butanoate, alanine, aspartate, and glutamate metabolism, and tricarboxylic acid (TCA) cycle processes. In conclusion, our study demonstrated that treatment with JTKY ameliorated poly(I:C)-induced pneumonia. The mechanism of action of JTKY may be associated with the inhibition of the inflammatory response, the reduction of oxidative stress, and the regulation of the synthesis and degradation of ketone bodies, TCA cycle, and metabolism of alanine, aspartate, glutamate, and butanoate processes in lung.
Collapse
Affiliation(s)
- Huantian Cui
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yuming Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bolun Yu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yulin Wu
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Gaijun Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junli Guo
- Hebei Hospital of Traditional Chinese Medicine, Hebei, China
| | - Junyu Luo
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
| | - Qin Li
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
- Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Xiaojuan Li
- Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, Zhejiang, China
| | - Wenju He
- Tianjin First Central Hospital, Tianjin, China
| | - Weibo Wen
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
- Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
- *Correspondence: Weibo Wen, ; Jiabao Liao, ; Dongqiang Wang,
| | - Jiabao Liao
- Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, Zhejiang, China
- *Correspondence: Weibo Wen, ; Jiabao Liao, ; Dongqiang Wang,
| | - Dongqiang Wang
- Tianjin First Central Hospital, Tianjin, China
- *Correspondence: Weibo Wen, ; Jiabao Liao, ; Dongqiang Wang,
| |
Collapse
|
40
|
Xia Y, Yao RQ, Zhao PY, Tao ZB, Zheng LY, Zhou HT, Yao YM, Song XM. Publication trends of research on COVID-19 and host immune response: A bibliometric analysis. Front Public Health 2022; 10:939053. [PMID: 36003630 PMCID: PMC9394856 DOI: 10.3389/fpubh.2022.939053] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/18/2022] [Indexed: 01/08/2023] Open
Abstract
IntroductionAs the first bibliometric analysis of COVID-19 and immune responses, this study will provide a comprehensive overview of the latest research advances. We attempt to summarize the scientific productivity and cooperation across countries and institutions using the bibliometric methodology. Meanwhile, using clustering analysis of keywords, we revealed the evolution of research hotspots and predicted future research focuses, thereby providing valuable information for the follow-up studies.MethodsWe selected publications on COVID-19 and immune response using our pre-designed search strategy. Web of Science was applied to screen the eligible publications for subsequent bibliometric analyses. GraphPad Prism 8.0, VOSviewer, and CiteSpace were applied to analyze the research trends and compared the contributions of countries, authors, institutions, and journals to the global publications in this field.ResultsWe identified 2,200 publications on COVID-19 and immune response published between December 1, 2019, and April 25, 2022, with a total of 3,154 citations. The United States (611), China (353), and Germany (209) ranked the top three in terms of the number of publications, accounting for 53.3% of the total articles. Among the top 15 institutions publishing articles in this area, four were from France, four were from the United States, and three were from China. The journal Frontiers in Immunology published the most articles (178) related to COVID-19 and immune response. Alessandro Sette (31 publications) from the United States were the most productive and influential scholar in this field, whose publications with the most citation frequency (3,633). Furthermore, the development and evaluation of vaccines might become a hotspot in relevant scope.ConclusionsThe United States makes the most indispensable contribution in this field in terms of publication numbers, total citations, and H-index. Although publications from China also take the lead regarding quality and quantity, their international cooperation and preclinical research need to be further strengthened. Regarding the citation frequency and the total number of published articles, the latest research progress might be tracked in the top-ranking journals in this field. By analyzing the chronological order of the appearance of retrieved keywords, we speculated that vaccine-related research might be the novel focus in this field.
Collapse
Affiliation(s)
- Yun Xia
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ren-qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Ren-qi Yao
| | - Peng-yue Zhao
- Department of General Surgery, First Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zheng-bo Tao
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Li-yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hui-ting Zhou
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yong-ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
- Yong-ming Yao
| | - Xue-min Song
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Xue-min Song
| |
Collapse
|
41
|
Liu L, Jing H, Wu X, Xiang M, Novakovic VA, Wang S, Shi J. The cross-talk of lung and heart complications in COVID-19: Endothelial cells dysfunction, thrombosis, and treatment. Front Cardiovasc Med 2022; 9:957006. [PMID: 35990983 PMCID: PMC9390946 DOI: 10.3389/fcvm.2022.957006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 01/08/2023] Open
Abstract
The pandemic respiratory illness SARS-CoV-2 has increasingly been shown to be a systemic disease that can also have profound impacts on the cardiovascular system. Although associated cardiopulmonary sequelae can persist after infection, the link between viral infection and these complications remains unclear. There is now a recognized link between endothelial cell dysfunction and thrombosis. Its role in stimulating platelet activation and thrombotic inflammation has been widely reported. However, the procoagulant role of microparticles (MPs) in COVID-19 seems to have been neglected. As membrane vesicles released after cell injury or apoptosis, MPs exert procoagulant activity mainly by exposing phosphatidylserine (PS) on their lipid membranes. It can provide a catalytic surface for the assembly of the prothrombinase complex. Therefore, inhibiting PS externalization is a potential therapeutic strategy. In this paper, we describe the pathophysiological mechanism by which SARS-CoV-2 induces lung and heart complications through injury of endothelial cells, emphasizing the procoagulant effect of MPs and PS, and demonstrate the importance of early antithrombotic therapy. In addition, we will detail the mechanisms underlying hypoxia, another serious pulmonary complication related to SARS-CoV-2-induced endothelial cells injury and discuss the use of oxygen therapy. In the case of SARS-CoV-2 infection, virus invades endothelial cells through direct infection, hypoxia, imbalance of the RAAS, and cytokine storm. These factors cause endothelial cells to release MPs, form MPs storm, and eventually lead to thrombosis. This, in turn, accelerates hypoxia and cytokine storms, forming a positive feedback loop. Given the important role of thrombosis in the disease, early antithrombotic therapy is an important tool for COVID-19. It may maintain normal blood circulation, accelerating the clearance of viruses, waning the formation of MPs storm, and avoiding disease progression.
Collapse
Affiliation(s)
- Langjiao Liu
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Haijiao Jing
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Mengqi Xiang
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Valerie A. Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States
| | - Shuye Wang
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- Shuye Wang
| | - Jialan Shi
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- *Correspondence: Jialan Shi ;
| |
Collapse
|
42
|
Moraes ECDS, Martins-Gonçalves R, da Silva LR, Mandacaru SC, Melo RM, Azevedo-Quintanilha I, Perales J, Bozza FA, Souza TML, Castro-Faria-Neto HC, Hottz ED, Bozza PT, Trugilho MRO. Proteomic Profile of Procoagulant Extracellular Vesicles Reflects Complement System Activation and Platelet Hyperreactivity of Patients with Severe COVID-19. Front Cell Infect Microbiol 2022; 12:926352. [PMID: 35937696 PMCID: PMC9354812 DOI: 10.3389/fcimb.2022.926352] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/20/2022] [Indexed: 01/08/2023] Open
Abstract
Background Extracellular vesicles (EVs) are a valuable source of biomarkers and display the pathophysiological status of various diseases. In COVID-19, EVs have been explored in several studies for their ability to reflect molecular changes caused by SARS-CoV-2. Here we provide insights into the roles of EVs in pathological processes associated with the progression and severity of COVID-19. Methods In this study, we used a label-free shotgun proteomic approach to identify and quantify alterations in EV protein abundance in severe COVID-19 patients. We isolated plasma extracellular vesicles from healthy donors and patients with severe COVID-19 by size exclusion chromatography (SEC). Then, flow cytometry was performed to assess the origin of EVs and to investigate the presence of circulating procoagulant EVs in COVID-19 patients. A total protein extraction was performed, and samples were analyzed by nLC-MS/MS in a Q-Exactive HF-X. Finally, computational analysis was applied to signify biological processes related to disease pathogenesis. Results We report significant changes in the proteome of EVs from patients with severe COVID-19. Flow cytometry experiments indicated an increase in total circulating EVs and with tissue factor (TF) dependent procoagulant activity. Differentially expressed proteins in the disease groups were associated with complement and coagulation cascades, platelet degranulation, and acute inflammatory response. Conclusions The proteomic data reinforce the changes in the proteome of extracellular vesicles from patients infected with SARS-CoV-2 and suggest a role for EVs in severe COVID-19.
Collapse
Affiliation(s)
- Emilly Caroline dos Santos Moraes
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Remy Martins-Gonçalves
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Luana Rocha da Silva
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Samuel Coelho Mandacaru
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Reynaldo Magalhães Melo
- Laboratory Protein Chemistry and Biochemistry and Laboratory of Gene Biology, Department of Cell Biology, University of Brasília, Brasília, Brazil
| | | | - Jonas Perales
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Fernando A. Bozza
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- D’Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Thiago Moreno Lopes Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Eugenio D. Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Patricia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Monique R. O. Trugilho
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Zhang LY, Huang LS, Yue YH, Fawaz R, Lim JK, Fan JG. Acute Hepatitis of Unknown Origin in Children: Early Observations from the 2022 Outbreak. J Clin Transl Hepatol 2022; 10:522-530. [PMID: 35836761 PMCID: PMC9240245 DOI: 10.14218/jcth.2022.00281] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 12/14/2022] Open
Abstract
Recent reports of acute hepatitis of unknown origin in previously healthy children have been increasing worldwide. The main characteristics of the affected children were jaundice and gastrointestinal symptoms. Their serum aminotransaminase levels were above 500 IU/L, with negative tests for hepatitis viruses A-E. By 31 May 2022, the outbreak had affected over 800 children under the age of 16 years in more than 40 countries, resulting in acute liver failure in approximately 10%, including at least 21 deaths and 38 patients requiring liver transplantation. There was still no confirmed cause or causes, although there were several different working hypotheses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), adenovirus serotype 41, or SARS-CoV-2 superantigen-mediated immune cell activation. Here, we review early observations of the 2022 outbreak which may inform diagnosis, treatment, and prevention in the context of an overlapping COVID-19 pandemic.
Collapse
Affiliation(s)
- Li-Ya Zhang
- Department of Infectious Disease, Xinhua Children’s Hospital, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Su Huang
- Department of Infectious Disease, Xinhua Children’s Hospital, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Hang Yue
- Department of Infectious Disease, Xinhua Children’s Hospital, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rima Fawaz
- Section of Pediatric Gastroenterology and Hepatology, Yale University School of Medicine, New Haven, CT, USA
| | - Joseph K. Lim
- Section of Digestive Diseases and Yale Liver Center, Yale University School of Medicine, New Haven, CT, USA
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| |
Collapse
|
44
|
Leng L, Ma J, Zhang PP, Xu SC, Li X, Jin Y, Cai J, Tang R, Zhao L, He ZC, Li MS, Zhang H, Zhou LR, Wu ZH, Li TR, Zhu YP, Wang YJ, Wu HB, Ping YF, Yao XH, Zhu CH, Guo HT, Tan LY, Liang ZY, Bian XW, Zhang SY. Spatial region-resolved proteome map reveals mechanism of COVID-19-associated heart injury. Cell Rep 2022; 39:110955. [PMID: 35679865 PMCID: PMC9135696 DOI: 10.1016/j.celrep.2022.110955] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/28/2022] [Accepted: 05/23/2022] [Indexed: 11/28/2022] Open
Abstract
Direct myocardial and vascular injuries due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection-driven inflammation is the leading cause of acute cardiac injury associated with coronavirus disease 2019 (COVID-19). However, in-depth knowledge of the injury characteristics of the heart affected by inflammation is lacking. In this study, using a quantitative spatial proteomics strategy that combines comparative anatomy, laser-capture microdissection, and histological examination, we establish a region-resolved proteome map of the myocardia and microvessels with obvious inflammatory cells from hearts of patients with COVID-19. A series of molecular dysfunctions of myocardia and microvessels is observed in different cardiac regions. The myocardia and microvessels of the left atrial are the most susceptible to virus infection and inflammatory storm, suggesting more attention should be paid to the lesion and treatment of these two parts. These results can guide in improving clinical treatments for cardiovascular diseases associated with COVID-19.
Collapse
Affiliation(s)
- Ling Leng
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Pei-Pei Zhang
- Department of Pathology, The First Hospital Affiliated to University of Science and Technology of China (USTC), Intelligent Pathology Institute, Division of Life Sciences and Medicine, USTC, Hefei, Anhui 230036, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China; Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Si-Chi Xu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiao Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Ye Jin
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jun Cai
- Department of Pathology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rui Tang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Lei Zhao
- Department of Pathology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Man-Sheng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Hui Zhang
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Liang-Rui Zhou
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zhi-Hong Wu
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Tian-Ran Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Yun-Ping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Basic Medical School, Anhui Medical University, Anhui 230032, China
| | - Yu-Jie Wang
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Hai-Bo Wu
- Department of Pathology, The First Hospital Affiliated to University of Science and Technology of China (USTC), Intelligent Pathology Institute, Division of Life Sciences and Medicine, USTC, Hefei, Anhui 230036, China
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Xiao-Hong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Chu-Hong Zhu
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hai-Tao Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Le-Yong Tan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Xiu-Wu Bian
- Department of Pathology, The First Hospital Affiliated to University of Science and Technology of China (USTC), Intelligent Pathology Institute, Division of Life Sciences and Medicine, USTC, Hefei, Anhui 230036, China; Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China.
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China; Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
45
|
Nawrot J, Gornowicz-Porowska J, Budzianowski J, Nowak G, Schroeder G, Kurczewska J. Medicinal Herbs in the Relief of Neurological, Cardiovascular, and Respiratory Symptoms after COVID-19 Infection A Literature Review. Cells 2022; 11:1897. [PMID: 35741026 PMCID: PMC9220793 DOI: 10.3390/cells11121897] [Citation(s) in RCA: 4] [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: 04/26/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 02/05/2023] Open
Abstract
COVID-19 infection causes complications, even in people who have had a mild course of the disease. The most dangerous seem to be neurological ailments: anxiety, depression, mixed anxiety-depressive (MAD) syndromes, and irreversible dementia. These conditions can negatively affect the respiratory system, circulatory system, and heart functioning. We believe that phytotherapy can be helpful in all of these conditions. Clinical trials confirm this possibility. The work presents plant materials (Valeriana officinalis, Melissa officinalis, Passiflora incarnata, Piper methysticum, Humulus lupulus, Ballota nigra, Hypericum perforatum, Rhodiola rosea, Lavandula officinalis, Paullinia cupana, Ginkgo biloba, Murraya koenigii, Crataegus monogyna and oxyacantha, Hedera helix, Polygala senega, Pelargonium sidoides, Lichen islandicus, Plantago lanceolata) and their dominant compounds (valeranon, valtrate, apigenin, citronellal, isovitexin, isoorientin, methysticin, humulone, farnesene, acteoside, hypericin, hyperforin, biapigenin, rosavidin, salidroside, linalool acetate, linalool, caffeine, ginkgolide, bilobalide, mihanimbine, epicatechin, hederacoside C,α-hederine, presegenin, umckalin, 6,7,8-trixydroxybenzopyranone disulfate, fumaroprotocetric acid, protolichesteric acid, aucubin, acteoside) responsible for their activity. It also shows the possibility of reducing post-COVID-19 neurological, respiratory, and cardiovascular complications, which can affect the functioning of the nervous system.
Collapse
Affiliation(s)
- Joanna Nawrot
- Department and Division of Practical Cosmetology and Skin Diseases Prophylaxis, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (J.G.-P.); (J.B.); (G.N.)
| | - Justyna Gornowicz-Porowska
- Department and Division of Practical Cosmetology and Skin Diseases Prophylaxis, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (J.G.-P.); (J.B.); (G.N.)
| | - Jaromir Budzianowski
- Department and Division of Practical Cosmetology and Skin Diseases Prophylaxis, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (J.G.-P.); (J.B.); (G.N.)
| | - Gerard Nowak
- Department and Division of Practical Cosmetology and Skin Diseases Prophylaxis, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (J.G.-P.); (J.B.); (G.N.)
| | - Grzegorz Schroeder
- Faculty of Chemistry, Adam Mickiewicz University in Poznan, Uniwersytetu 5, Poznanskiego 8, 61-614 Poznan, Poland; (G.S.); (J.K.)
| | - Joanna Kurczewska
- Faculty of Chemistry, Adam Mickiewicz University in Poznan, Uniwersytetu 5, Poznanskiego 8, 61-614 Poznan, Poland; (G.S.); (J.K.)
| |
Collapse
|