1
|
Mei W, Zhu Y, Nie Z, Fang C, Zhang Z, Qiu H, Bai X, Pan L. Retrospective analysis of venous thromboembolism, arterial thromboembolism, and microthrombosis incidence at a single center during the COVID-19 pandemic. Medicine (Baltimore) 2024; 103:e39915. [PMID: 39465786 PMCID: PMC11479453 DOI: 10.1097/md.0000000000039915] [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: 06/19/2024] [Accepted: 09/13/2024] [Indexed: 10/29/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pneumonia caused by severe acute respiratory syndrome coronavirus 2 has posed a significant threat to global health since its outbreak in early 2020. Various thrombotic complications have been reported in COVID-19 cases. This study aims to investigate the incidence of various types of thromboses during the COVID-19 pandemic, and explore the potential correlation between the number of COVID-19 infections and occurrence of thrombosis. The present retrospective, single-center study recorded the number of new cases with various types of thromboembolism among all hospital admissions between 2019 and 2023 in a single center. The correlation between the number of total thromboembolism cases and number of inpatients who tested positive for COVID-19 was assessed using Spearman rank correlation coefficient. A total of 3926 thromboembolism cases were recorded in men, while 3657 cases were reported in women. The total number of new thromboembolism cases among all inpatients annually increased from 2019 to 2022, and reached a peak during the "full liberalization" period from December 2022 to January 2023. The most substantial increase was observed in microthrombosis cases, followed by venous and arterial thrombosis cases. In addition, thromboembolism primarily occurred in the lower extremities, followed by the abdomen. The fewest cases were observed in the upper extremities. Furthermore, the correlation coefficient between the total number of thromboembolism cases and number of COVID-19-positive patients between January 2020 and June 2023 was 0.501 (P = .001). The number of thromboembolism cases among all inpatients was correlated to the number of COVID-19-positive patients during the pandemic. Early thromboembolism prophylaxis and thrombotic complications monitoring should be considered for hospitalized patients, when necessary.
Collapse
Affiliation(s)
- Wenjun Mei
- Anqing Municipal Hospital, Anqing City, Anhui Province, China
| | - Yuefeng Zhu
- Sir Run Run Shaw Hospital, Hangzhou, Zhejiang Province, China
| | - Zongjie Nie
- Anqing Municipal Hospital, Anqing City, Anhui Province, China
| | - Canjun Fang
- Anqing Municipal Hospital, Anqing City, Anhui Province, China
| | - Zheng Zhang
- Anqing Municipal Hospital, Anqing City, Anhui Province, China
| | - Haijun Qiu
- Anqing Municipal Hospital, Anqing City, Anhui Province, China
| | - Xia Bai
- Anqing Municipal Hospital, Anqing City, Anhui Province, China
| | - Lisheng Pan
- Anqing Municipal Hospital, Anqing City, Anhui Province, China
| |
Collapse
|
2
|
Le DCP, Bui HT, Vo QD. Endovascular Intervention for Acute Superior Mesenteric Artery Occlusion Following COVID-19 Pneumonia: Two Case Reports. Interv Cardiol 2024; 19:e17. [PMID: 39309299 PMCID: PMC11413984 DOI: 10.15420/icr.2024.09] [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: 03/25/2024] [Accepted: 07/14/2024] [Indexed: 09/25/2024] Open
Abstract
COVID-19 patients may experience acute mesenteric ischaemia. Identifying acute mesenteric ischaemia is challenging, particularly as initial symptoms are often vague and easily overlooked. Early detection and immediate intervention to restore blood flow can prevent these severe consequences. Presented in this report are two cases of superior mesenteric artery (SMA) thrombosis following severe acute respiratory syndrome coronavirus 2 infection. CT scans demonstrated SMA thrombosis in both patients, with no evidence of bowel necrosis. Endovascular intervention with self-expanding stent placement was performed after angiographic confirmation of the diagnosis. At 6-month follow-up, both patients remained asymptomatic on dual antiplatelet therapy. Atypical gastrointestinal manifestations in COVID-19 patients should raise suspicion for uncommon complications, such as SMA thrombosis. For SMA occlusion without associated bowel necrosis, endovascular therapy represents a viable treatment approach.
Collapse
Affiliation(s)
- Duy Cao Phuong Le
- Faculty of Medicine, Nguyen Tat Thanh UniversityHo Chi Minh City Vietnam
- Department of Cardiovascular Intervention, Nguyen Tri Phuong HospitalHo Chi Minh City, Vietnam
| | - Hoa The Bui
- Department of Cardiovascular Intervention, Nguyen Tri Phuong HospitalHo Chi Minh City, Vietnam
| | - Quan Duy Vo
- Faculty of Medicine, Nguyen Tat Thanh UniversityHo Chi Minh City Vietnam
- Department of Cardiovascular Intervention, Nguyen Tri Phuong HospitalHo Chi Minh City, Vietnam
| |
Collapse
|
3
|
Han SH, Ockerman K, Kirchmier M, Mardourian M, Bryan J, Cox E, Chim H, Spiguel L, Momeni A, Sorice-Virk S. Thrombotic Consequences of COVID-19 Infection on Microsurgical Reconstruction. Microsurgery 2024; 44:e31219. [PMID: 39207212 DOI: 10.1002/micr.31219] [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: 12/14/2023] [Revised: 06/12/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Evidence has shown increased morbidity and mortality for patients with COVID-19 infection within 7 weeks of surgery. However, no studies have specifically investigated the effects of COVID-19 in microsurgical outcomes. This study evaluated thrombotic and overall complications after free tissue transfer for a variety of indications in patients with and without previous COVID-19 infection. METHODS A retrospective cohort study was performed in adult patients with or without a history of COVID-19 infection who underwent microsurgical reconstruction between 2017 and 2022. Patients with a history of COVID-19 infection were matched to controls based on age, gender, race, body mass index, history of diabetes, coronary artery disease, hypertension, Caprini score, tobacco use, and flap indication. RESULTS From 2017 to 2022, 35 patients had a documented history of COVID-19. Matched case analysis determined a 4.8 times increased odds ratio of postoperative complications in the COVID-19 group compared with controls (p = 0.002). Significantly, more patients with COVID-19 experienced total or partial flap loss and anastomotic issues (COVID-19: 7/35, Control: 0/35; p < 0.001). There was no significant difference in incidence of VTE (COVID-19: 1/35, Control: 0/35; p = 0.493). Of note, 62.9% of the COVID-19 group were discharged on anticoagulants (versus 14.3% in the control group [p < 0.001]). CONCLUSION COVID-19 has dire, long-lasting effects on virtually every organ system, chief among them, the microcirculation. Further studies are needed to fully determine the extent and influence of COVID-19 on complex procedures such as free tissue transfer and how to optimize the screening, workup, and postoperative care to guard against the associated thrombotic consequences.
Collapse
Affiliation(s)
- Sabrina H Han
- Division of Plastic and Reconstructive Surgery, University of Chicago, Chicago, Illinois, USA
| | - Kyle Ockerman
- Division of Plastic and Reconstructive Surgery, University of Chicago, Chicago, Illinois, USA
| | - Matthew Kirchmier
- Division of Plastic and Reconstructive Surgery, University of Chicago, Chicago, Illinois, USA
| | - Markos Mardourian
- Division of Plastic and Reconstructive Surgery, University of Colorado, Aurora, Colorado, USA
| | - Jaimie Bryan
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, California, USA
| | - Elizabeth Cox
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, California, USA
| | - Harvey Chim
- Division of Plastic and Reconstructive Surgery, University of Florida, Gainesville, Florida, USA
| | - Lisa Spiguel
- Division of Surgical Oncology, University of Florida, Gainesville, Florida, USA
| | - Arash Momeni
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, California, USA
| | - Sarah Sorice-Virk
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, California, USA
| |
Collapse
|
4
|
Kenney D, O’Connell AK, Tseng AE, Turcinovic J, Sheehan ML, Nitido AD, Montanaro P, Gertje HP, Ericsson M, Connor JH, Vrbanac V, Crossland NA, Harly C, Balazs AB, Douam F. A Novel Human Extravascular Monocyte Subset with Antiviral Functions Is Crucial for Resolving Lung Tissue Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.583965. [PMID: 38496468 PMCID: PMC10942442 DOI: 10.1101/2024.03.08.583965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The recurring emergence of novel respiratory viruses has highlighted our poor understanding of the human immune mechanisms governing the resolution of lung infection in an immunologically naïve context. Using SARS-CoV-2 as a prototypical emerging respiratory virus, we leveraged mice co-engrafted with a genetically matched fetal lung xenograft (fLX) and a human immune system (BLT-L mice) to investigate such mechanisms. While BLT-L mice effectively resolve SARS-CoV-2 infection following acute viral replication in fLX, viral clearance is robustly abrogated through systemic depletion of CD4+, but not CD3+ or CD8+ cells, resulting in persistent infection. Leveraging single-cell transcriptomics to uncover the CD4-expressing subsets driving infection resolution, we identified a novel subset of lung extravascular inflammatory monocytes (ExiMO) with antiviral functions. ExiMO are the dominant CD163-expressing myeloid population emerging in fLX upon acute infection and derive from recruited circulating CD4+ monocytes. They are highly enriched in viral RNA and elicit a robust antiviral response before vanishing from tissues when infection resolves. Notably, systemic CD4+ cell depletion results in impaired recruitment of CD163+ cells into fLX and leads to a state of immune tolerance and chronic infection defined by the absence of ExiMO antiviral responses. Together, our study uncovers ExiMO as major sentinels driving SARS-CoV-2 infection resolution in human lung tissues without pre-existing immunity. This work expands our understanding of lung extravascular monocytes and unravels novel facets of the cellular determinants governing our vulnerability to viral respiratory pathogens.
Collapse
Affiliation(s)
- Devin Kenney
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Aoife K. O’Connell
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Anna E. Tseng
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jacquelyn Turcinovic
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Maegan L. Sheehan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Adam D. Nitido
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Paige Montanaro
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Hans P. Gertje
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Maria Ericsson
- Electron Microscopy Core Facility, Harvard Medical School, Boston, MA, USA
| | - John H. Connor
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | | | - Nicholas A. Crossland
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Christelle Harly
- Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France
- LabEx IGO 'Immunotherapy, Graft, Oncology', Nantes, France
- These authors contributed equally to the work
| | - Alejandro B. Balazs
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally to the work
| | - Florian Douam
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- These authors contributed equally to the work
- Lead contact
| |
Collapse
|
5
|
Lemon NM, Taylor LK, Rech MA, Nguyen Q, Matthews GJ, Lew G, Lovett S. Utility of D-dimer in predicting pulmonary embolism in patients with COVID-19 presenting to the emergency department. J Am Coll Emerg Physicians Open 2024; 5:e13237. [PMID: 39027350 PMCID: PMC11255020 DOI: 10.1002/emp2.13237] [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: 09/22/2023] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Objectives While our understanding of coronavirus disease 2019 (COVID-19) has evolved, uncertainty remains regarding utility of previously established pulmonary embolism (PE) screening guidelines in patients with COVID-19. Many studies have investigated the efficacy of D-dimer (DD) screenings for patients with COVID-19 admitted to inpatient services, but few have evaluated patients in the emergency department (ED). The purpose of this study was to investigate utility of DD threshold for PE screening in patients with COVID-19 presenting to the ED. Methods This was a retrospective, multicenter cohort including patients presenting to three EDs between March 1, 2020 and February 1, 2021 who tested positive for COVID-19 during ED visit or in 60 days prior to presentation and had DD ordered in ED. Patients were grouped by those who underwent computed tomography pulmonary angiogram (CTPA) to evaluate for PE and those who did not, and descriptive statistics were performed. Those who underwent CTPA were further divided into PE-positive and PE-negative groups. The discriminative ability of DD in predicting PE in patients with COVID-19 was analyzed using the receiver operating characteristic (ROC) curve. Results A total of 570 patients with COVID-19 were included in the study, of which 107 underwent CTPA to evaluate for PE. History of diabetes, elevated glucose, elevated lactate dehydrogenase, elevated white blood cell count, elevated platelets, elevated respiratory rate, and lower temperature were associated with increased risk for PE. Compared to those without PE, patients with PE were significantly more likely to be hospitalized (100% vs. 82%, p = 0.020) and admitted to the ICU (64% vs. 24%, p = 0.002). Those with PE had a significantly higher median DD value (21,177 ng/mL) compared to PE-negative group (952 ng/mL, p < 0.001). The ROC curve for DD in predicting PE had an area under the curve of 0.91 (95% confidence interval [0.84, 0.98]). In our study population, the optimal DD threshold for predicting PE was 1815 ng/mL (sensitivity 93% and specificity 80%). A conservative threshold of 1089 ng/mL could be used with sensitivity 100% and specificity 58%. Conclusion DD is often elevated in patients with COVID-19, regardless of PE. While the classically used DD cutoff is 500 ng/mL, our study demonstrated a threshold of 1089 ng/mL safely predicted PE in patients with COVID-19 .
Collapse
Affiliation(s)
- Natalie M. Lemon
- Department of Emergency MedicineUniversity of Chicago Medical CenterChicagoIllinoisUSA
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Luke K. Taylor
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Megan A. Rech
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
- Loyola University Medical CenterChicagoIllinoisUSA
- Center of Innovation for Complex Chronic HealthcareEdward Hines VA HospitalHinesIllinoisUSA
| | - Quang Nguyen
- Department of Statistics and Data ScienceCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Center for Data Science and ConsultingLoyola University ChicagoChicagoIllinoisUSA
| | - Gregory J. Matthews
- Center for Data Science and ConsultingLoyola University ChicagoChicagoIllinoisUSA
- Department of Mathematics and StatisticsLoyola University ChicagoChicagoIllinoisUSA
| | - George Lew
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
- Loyola University Medical CenterChicagoIllinoisUSA
| | - Shannon Lovett
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
- Loyola University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
6
|
Recchia Luciani G, Barilli A, Visigalli R, Dall’Asta V, Rotoli BM. Cytokines from SARS-CoV-2 Spike-Activated Macrophages Hinder Proliferation and Cause Cell Dysfunction in Endothelial Cells. Biomolecules 2024; 14:927. [PMID: 39199315 PMCID: PMC11353037 DOI: 10.3390/biom14080927] [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: 06/18/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Endothelial dysfunction plays a central role in the severity of COVID-19, since the respiratory, thrombotic and myocardial complications of the disease are closely linked to vascular endothelial damage. To address this issue, we evaluate here the effect of conditioned media from spike S1-activated macrophages (CM_S1) on the proliferation of human umbilical endothelial cells (HUVECs), focusing on the specific role of interleukin-1-beta (IL-1β), interleukin-6 (IL-6), interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). Results obtained demonstrate that the incubation with CM_S1 for 72 h hinders endothelial cell proliferation and induces signs of cytotoxicity. Comparable results are obtained upon exposure to IFN-γ + TNF-α, which are thus postulated to play a pivotal role in the effects observed. These events are associated with an increase in p21 protein and a decrease in Rb phosphorylation, as well as with the activation of IRF-1 and NF-kB transcription factors. Overall, these findings further sustain the pivotal role of a hypersecretion of inflammatory cytokines as a trigger for endothelial activation and injury in the immune-mediated effects of COVID-19.
Collapse
Affiliation(s)
| | | | | | - Valeria Dall’Asta
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (G.R.L.); (A.B.); (R.V.); (B.M.R.)
| | | |
Collapse
|
7
|
Li Y, Yang D, Kang J, Cao Y, Cui L, Liu F. COVID-19 and the risk of acute cardiovascular diseases: a two-sample Mendelian randomization study. BMC Cardiovasc Disord 2024; 24:389. [PMID: 39068390 PMCID: PMC11282648 DOI: 10.1186/s12872-024-04066-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Evidence suggests that coronavirus disease 2019 (COVID-19) is associated with the risk of cardiovascular diseases (CVDs). However, the results are inconsistent, and the causality remains to be established. We aimed to investigate the potential causal relationship between COVID-19 and CVDs by using two-sample Mendelian randomization (MR) analysis. METHODS Summary-level data for COVID-19 and CVDs including myocarditis, heart failure (HF), acute myocardial infarction (AMI), arrhythmia and venous thromboembolism (VTE) were obtained from the IEU OpenGWAS project, a public genome-wide association study (GWAS). Single nucleotide polymorphisms (SNPs) were used as instrumental variables. Five complementary MR methods were performed, including inverse variance weighted (IVW), MR-Egger, weighted median, weighted mode and simple mode methods. IVW method was considered as the primary approach. Besides, sensitivity analyses, including Cochran's Q test, MR-Egger intercept test, and leave-one-out analysis, were performed to evaluate the robustness of the results. RESULTS According to the IVW results, our MR study indicated that genetically predicted COVID-19 was not causally connected with the risk of CVDs [myocarditis: odds ratio (OR) = 1.407, 95% confidence interval (CI) = 0.761-2.602, p-value = 0.277; HF: OR = 1.180, 95% CI = 0.980-1.420, p-value = 0.080; AMI: OR = 1.002, 95% CI = 0.998-1.005, p-value = 0.241; arrhythmia: OR = 0.865, 95% CI = 0.717-1.044, p-value = 0.132; VTE: OR = 1.013, 95% CI = 0.997-1.028, p-value = 0.115]. The supplementary MR methods showed similar results. Sensitivity analyses suggested that the causal estimates were robust. CONCLUSION This two-sample MR analysis did not provide sufficient evidence for a causal relationship between COVID-19 and the risk of acute CVDs, which may provide new insights into the prevention of acute CVDs in COVID-19 patients.
Collapse
Affiliation(s)
- Yuling Li
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
- Emergency Department, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | | | - Jian Kang
- Emergency Department, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
- Phase I Clinical Trials Center, The First Hospital, China Medical University, 518 North Chuangxin Road, Baita Street, Hunnan District, Shenyang, Liaoning, 110102, China.
| |
Collapse
|
8
|
Krivdić Dupan Z, Periša V, Suver Stević M, Mihalj M, Tolušić Levak M, Guljaš S, Salha T, Loinjak D, Kos M, Šapina M, Canjko I, Šambić Penc M, Štefančić M, Nešković N. The Impact of Pentraxin 3 Serum Levels and Angiotensin-Converting Enzyme Polymorphism on Pulmonary Infiltrates and Mortality in COVID-19 Patients. Biomedicines 2024; 12:1618. [PMID: 39062191 PMCID: PMC11275229 DOI: 10.3390/biomedicines12071618] [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: 07/06/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
OBJECTIVES The aim of this study was to examine the impact of the pentraxin 3 (PTX3) serum level and angiotensin-converting enzyme (ACE) gene insertion/deletion (I/D) polymorphism on the severity of radiographic pulmonary infiltrates and the clinical outcomes of COVID-19. METHODS The severity of COVID-19 pulmonary infiltrates was evaluated within a week of admission by analyzing chest X-rays (CXR) using the modified Brixia (MBrixa) scoring system. The insertion (I)/deletion (D) polymorphism of the ACE gene and the serum levels of PTX3 were determined for all patients included in the study. RESULTS This study included 80 patients. Using a cut-off serum level of PTX3 ≥ 2.765 ng/mL, the ROC analysis (AUC 0.871, 95% CI 0.787-0.954, p < 0.001) showed a sensitivity of 85.7% and specificity of 78.8% in predicting severe MBrixa scores. Compared to ACE I/I polymorphism, D/D polymorphism significantly increased the risk of severe CXR infiltrates, OR 7.7 (95% CI: 1.9-30.1), and p = 0.002. Significant independent predictors of severe CXR infiltrates include hypertension (OR 7.71), PTX3 (OR 1.20), and ACE D/D polymorphism (OR 18.72). Hypertension (OR 6.91), PTX3 (OR 1.47), and ACE I/I polymorphism (OR 0.09) are significant predictors of poor outcomes. CONCLUSION PTX3 and ACE D/D polymorphism are significant predictors of the severity of COVID-19 pneumonia. PTX3 is a significant predictor of death.
Collapse
Affiliation(s)
- Zdravka Krivdić Dupan
- Department of Radiology, Osijek University Hospital, 31000 Osijek, Croatia
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Vlatka Periša
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Hematology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Mirjana Suver Stević
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Transfusion Medicine, Osijek University Hospital, 31000 Osijek, Croatia
| | - Martina Mihalj
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Dermatology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Maja Tolušić Levak
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Dermatology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Silva Guljaš
- Department of Radiology, Osijek University Hospital, 31000 Osijek, Croatia
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Tamer Salha
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Dental Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Domagoj Loinjak
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Internal Medicine, Osijek University Hospital, 31000 Osijek, Croatia
| | - Martina Kos
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Pediatrics, Osijek University Hospital, 31000 Osijek, Croatia
| | - Matej Šapina
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Pediatrics, Osijek University Hospital, 31000 Osijek, Croatia
| | - Ivana Canjko
- Department of Radiotherapy and Oncology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Mirela Šambić Penc
- Department of Radiotherapy and Oncology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Marin Štefančić
- Department of Radiology, National Memorial Hospital Vukovar, 32000 Vukovar, Croatia
| | - Nenad Nešković
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- International Medical Center Priora, 31431 Cepin, Croatia
| |
Collapse
|
9
|
Maurício AB, Cavalcante AMRZ, de Sá ES, Bruni LG, Vieira LGD, Costa A, França LD, Lopes MVDO, de Barros ALBL, da Silva VM. Accuracy of the defining characteristics of respiratory nursing diagnoses in patients with COVID-19. Int J Nurs Knowl 2024. [PMID: 38951045 DOI: 10.1111/2047-3095.12481] [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: 04/24/2024] [Accepted: 06/12/2024] [Indexed: 07/03/2024]
Abstract
OBJECTIVE To analyze the accuracy of the defining characteristics of four respiratory nursing diagnoses (ND) in patients with COVID-19 and on oxygen therapy. METHODS This is a cross-sectional study conducted in four Brazilian public hospitals in two regions of the country. A total of 474 patients with COVID-19 receiving oxygen therapy were assessed. Latent-adjusted class analysis with random effects was used to establish the sensitivity (Se) and specificity (Sp) of the defining characteristics evaluated for each ND. RESULTS Among the ND that constituted the study (impaired spontaneous ventilatory, impaired gas exchange, ineffective airway clearance, and dysfunctional ventilatory weaning response), the following defining characteristics had the highest simultaneous Se and Sp (>0.8): decrease in tidal volume, confusion, irritability, dyspnea, decreased breath sounds, orthopnea, impaired ability to cooperate and respond to coaching, and decrease in the level of consciousness. CONCLUSIONS Recognizing the clinical signs that predict respiratory ND in patients affected by COVID-19 can contribute to the nurse's accurate diagnostic inference and designate the appropriate nursing interventions to achieve the desired results and avoid complications.
Collapse
Affiliation(s)
| | | | | | | | | | - Adriana Costa
- Paulista Nursing School, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
10
|
Abramowitz BR, Coles M, Aytaman A, Chander-Roland B, DiLeo DA. Simultaneous portal vein thrombosis and splenic vein thrombosis in a COVID-19 patient: A case report and review of literature. World J Clin Cases 2024; 12:3561-3566. [PMID: 38983408 PMCID: PMC11229906 DOI: 10.12998/wjcc.v12.i18.3561] [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: 01/29/2024] [Revised: 04/09/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND It is well-described that the coronavirus disease 2019 (COVID-19) infection is associated with an increased risk of thrombotic complications. While there have been many cases of pulmonary emboli and deep vein thrombosis in these patients, reports of COVID-19 associated portal vein thrombosis (PVT) have been uncommon. We present a unique case of concomitant PVT and splenic artery thrombosis in a COVID-19 patient. CASE SUMMARY A 77-year-old-male with no history of liver disease presented with three days of left-sided abdominal pain. One week earlier, the patient was diagnosed with mildly symptomatic COVID-19 and was treated with nirmatrelvir/ritonavir. Physical exam revealed mild right and left lower quadrant tenderness, but was otherwise unremarkable. Significant laboratory findings included white blood cell count 12.5 K/μL, total bilirubin 1.6 mg/dL, aminoaspartate transferase 40 U/L, and alanine aminotransferase 61 U/L. Computed tomography of the abdomen and pelvis revealed acute PVT with thrombus extending from the distal portion of the main portal vein into the right and left branches. Also noted was a thrombus within the distal portion of the splenic artery with resulting splenic infarct. Hypercoagulable workup including prothrombin gene analysis, factor V Leiden, cardiolipin antibody, and JAK2 mutation were all negative. Anticoagulation with enoxaparin was initiated, and the patient's pain improved. He was discharged on apixaban. CONCLUSION It is quite uncommon for PVT to present simultaneously with an arterial thrombotic occlusion, as in the case of our patient. Unusual thrombotic manifestations are classically linked to hypercoagulable states including malignancy and hereditary and autoimmune disorders. Viral infections such as Epstein-Barr virus, cytomegalovirus, viral hepatitis, and COVID-19 have all been found to increase the risk of splanchnic venous occlusions, including PVT. In our patient, prompt abdominal imaging led to early detection of thrombus, early treatment, and an excellent outcome. This case is unique in that it is the second known case within the literature of simultaneous PVT and splenic artery thrombosis in a COVID-19 patient.
Collapse
Affiliation(s)
- Binyamin Ravina Abramowitz
- Department of Gastroenterology and Hepatology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Michael Coles
- Department of Gastroenterology and Hepatology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Ayse Aytaman
- Department of Gastroenterology and Hepatology, Brooklyn Campus of the Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY 11209, United States
| | - Bani Chander-Roland
- Department of Gastroenterology and Hepatology, Brooklyn Campus of the Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY 11209, United States
| | - Daniel Anthony DiLeo
- Department of Gastroenterology and Hepatology, Brooklyn Campus of the Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY 11209, United States
| |
Collapse
|
11
|
Nechita L, Niculet E, Baroiu L, Balta AAS, Nechita A, Voinescu DC, Manole C, Busila C, Debita M, Tatu AL. Acute Myocardial Infarction in COVID-19 Patients-A Review of Literature Data and Two-Case Report Series. J Clin Med 2024; 13:2936. [PMID: 38792477 PMCID: PMC11121956 DOI: 10.3390/jcm13102936] [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: 04/23/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Background/Objectives: The newly emergent COVID-19 pandemic involved primarily the respiratory system and had also major cardiovascular system (CVS) implications, revealed by acute myocardial infarction (AMI), arrhythmias, myocardial injury, and thromboembolism. CVS involvement is done through main mechanisms-direct and indirect heart muscle injury, with high mortality rates, worse short-term outcomes, and severe complications. AMI is the echo of myocardial injury (revealed by increases in CK, CK-MB, and troponin serum markers-which are taken into consideration as possible COVID-19 risk stratification markers). When studying myocardial injury, physicians can make use of imaging studies, such as cardiac MRI, transthoracic (or transesophageal) echocardiography, coronary angiography, cardiac computed tomography, and nuclear imaging (which have been used in cases where angiography was not possible), or even endomyocardial biopsy (which is not always available or feasible). Two-case-series presentations: We present the cases of two COVID-19 positive male patients who were admitted into the Clinical Department of Cardiology in "Sfântul Apostol Andrei" Emergency Clinical Hospital of Galați (Romania), who presented with acute cardiac distress symptoms and have been diagnosed with ST elevation AMI. The patients were 82 and 57 years old, respectively, with moderate and severe forms of COVID-19, and were diagnosed with anteroseptal left ventricular AMI and extensive anterior transmural left ventricular AMI (with ventricular fibrillation at presentation), respectively. The first patient was a non-smoker and non-drinker with no associated comorbidities, and was later discharged, while the second one died due to AMI complications. Conclusions: From this two-case series, we extract the following: old age alone is not a significant risk factor for adverse outcomes in COVID-19-related CVS events, and that the cumulative effects of several patient-associated risk factors (be it either for severe forms of COVID-19 and/or acute cardiac injury) will most probably lead to poor patient prognosis (death). At the same time, serum cardiac enzymes, dynamic ECG changes, along with newly developed echocardiographic modifications are indicators for poor prognosis in acute cardiac injury in COVID-19 patients with acute myocardial injury, regardless of the presence of right ventricular dysfunction (due to pulmonary hypertension).
Collapse
Affiliation(s)
- Luiza Nechita
- Doctoral School of Biomedical Sciences, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.N.); (A.A.S.B.)
| | - Elena Niculet
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galați, Romania
- ‘Sf. Apostol Andrei’ Clinical Emergency County Hospital, 800578 Galati, Romania; (D.C.V.); (C.M.)
| | - Liliana Baroiu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (A.N.); (C.B.); (M.D.); (A.L.T.)
- ‘Sf. Cuv. Parascheva’ Clinical Hospital of Infectious Diseases, 800179 Galati, Romania
| | | | - Aurel Nechita
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (A.N.); (C.B.); (M.D.); (A.L.T.)
- ‘Sf. Ioan’ Clinical Hospital for Children, 800487 Galati, Romania
| | - Doina Carina Voinescu
- ‘Sf. Apostol Andrei’ Clinical Emergency County Hospital, 800578 Galati, Romania; (D.C.V.); (C.M.)
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (A.N.); (C.B.); (M.D.); (A.L.T.)
| | - Corina Manole
- ‘Sf. Apostol Andrei’ Clinical Emergency County Hospital, 800578 Galati, Romania; (D.C.V.); (C.M.)
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (A.N.); (C.B.); (M.D.); (A.L.T.)
| | - Camelia Busila
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (A.N.); (C.B.); (M.D.); (A.L.T.)
- ‘Sf. Ioan’ Clinical Hospital for Children, 800487 Galati, Romania
| | - Mihaela Debita
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (A.N.); (C.B.); (M.D.); (A.L.T.)
- ‘Sf. Cuv. Parascheva’ Clinical Hospital of Infectious Diseases, 800179 Galati, Romania
| | - Alin Laurentiu Tatu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (A.N.); (C.B.); (M.D.); (A.L.T.)
- ‘Sf. Cuv. Parascheva’ Clinical Hospital of Infectious Diseases, 800179 Galati, Romania
| |
Collapse
|
12
|
Bilehjani E, Fakhari S, Farzin H, Tajlil A, Nader ND. Diagnosis and treatment of cardiovascular manifestations of COVID-19: a narrative review. Acta Cardiol 2024; 79:267-273. [PMID: 37606350 DOI: 10.1080/00015385.2023.2246200] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) was the main pathogen in the COVID-19 pandemic. This viral infection has been associated with several respiratory and non-respiratory complications contributing to a higher mortality rate, especially in patients with underlying heart diseases worldwide. Once considered a respiratory tract disease, it is now well-known that COVID-19 patients may experience a wide range of cardiac manifestations. Because of its remarkable direct and indirect effects on the cardiovascular system, herein, we examined the published literature that studied the hypothetical mechanisms of injury, manifestations, and diagnostic modalities, including changes in molecular biomarkers with a predictive value in the prognostication of the disease, as well as emerging evidence regarding the long-term cardiac complications of the disease.
Collapse
Affiliation(s)
- Eissa Bilehjani
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Fakhari
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haleh Farzin
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezou Tajlil
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Nader D Nader
- Departments of Anesthesiology and Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| |
Collapse
|
13
|
Gheban-Roșca IA, Gheban BA, Pop B, Mironescu DC, Siserman VC, Jianu EM, Drugan T, Bolboacă SD. Immunohistochemical and Morphometric Analysis of Lung Tissue in Fatal COVID-19. Diagnostics (Basel) 2024; 14:914. [PMID: 38732328 PMCID: PMC11082993 DOI: 10.3390/diagnostics14090914] [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: 03/15/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The primary targets of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the lungs are type I pneumocytes, macrophages, and endothelial cells. We aimed to identify lung cells targeted by SARS-CoV-2 using viral nucleocapsid protein staining and morphometric features on patients with fatal COVID-19. We conducted a retrospective analysis of fifty-one autopsy cases of individuals who tested positive for SARS-CoV-2. Demographic and clinical information were collected from forensic reports, and lung tissue was examined for microscopic lesions and the presence of specific cell types. Half of the evaluated cohort were older than 71 years, and the majority were male (74.5%). In total, 24 patients presented diffuse alveolar damage (DAD), and 50.9% had comorbidities (56.9% obesity, 33.3% hypertension, 15.7% diabetes mellitus). Immunohistochemical analysis showed a similar pattern of infected macrophages, infected type I pneumocytes, and endothelial cells, regardless of the presence of DAD (p > 0.5). The immunohistochemical reactivity score (IRS) was predominantly moderate but without significant differences between patients with and without DAD (p = 0.633 IRS for type I pneumocytes, p = 0.773 IRS for macrophage, and p = 0.737 for IRS endothelium). The nucleus/cytoplasm ratio shows lower values in patients with DAD (median: 0.29 vs. 0.35), but the difference only reaches a tendency for statistical significance (p = 0.083). Our study confirms the presence of infected macrophages, type I pneumocytes, and endothelial cells with a similar pattern in patients with and without diffuse alveolar damage.
Collapse
Affiliation(s)
- Ioana-Andreea Gheban-Roșca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
- Clinical Hospital for Infectious Diseases, 400348 Cluj-Napoca, Romania
| | - Bogdan-Alexandru Gheban
- County Emergency Clinical Hospital, 400006 Cluj-Napoca, Romania
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Bogdan Pop
- The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania;
- Department of Anatomic Pathology, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Daniela-Cristina Mironescu
- Forensic Institute, 400006 Cluj-Napoca, Romania; (D.-C.M.); (V.C.S.)
- Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Vasile Costel Siserman
- Forensic Institute, 400006 Cluj-Napoca, Romania; (D.-C.M.); (V.C.S.)
- Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Elena Mihaela Jianu
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Tudor Drugan
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
| | - Sorana D. Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.-A.G.-R.); (T.D.)
| |
Collapse
|
14
|
de Sá ES, Maurício AB, Bruni LG, Vieira LGD, Santos VB, Cavalcante AMRZ, de Barros ALBL, da Silva VM. Content analysis of NOC outcomes related to mechanical ventilation in people with COVID-19. Rev Esc Enferm USP 2024; 58:e20230343. [PMID: 38587402 PMCID: PMC11000577 DOI: 10.1590/1980-220x-reeusp-2023-0343en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/16/2024] [Indexed: 04/09/2024] Open
Abstract
OBJECTIVE To analyze the evidence of content validity of the Nursing Outcomes "Mechanical Ventilation Response: Adult" and "Mechanical Ventilation Weaning Response: Adult", for patients with severe COVID-19. METHOD Methodological study developed in two stages: literature review to construct the definitions of the indicators and analysis of the evidence of content validity of the nursing outcomes by a focus group. RESULTS All the conceptual and operational definitions developed for the 56 indicators were considered clear and precise. However, 17 indicators were excluded because they were deemed not to be relevant. The definitions of the magnitudes for 17 indicators of the Nursing Outcome "Mechanical Ventilation Response: Adult" and 22 indicators "Mechanical Ventilation Weaning Response: Adult" were thus constructed. CONCLUSION The development of definitions and validation by experts makes the use of these outcomes and their indicators more understandable and precise, favoring their use in clinical practice and providing greater detail in assessment and recording.
Collapse
|
15
|
Mone P, Jankauskas SS, Manzi MV, Gambardella J, Coppola A, Kansakar U, Izzo R, Fiorentino G, Lombardi A, Varzideh F, Sorriento D, Trimarco B, Santulli G. Endothelial Extracellular Vesicles Enriched in microRNA-34a Predict New-Onset Diabetes in Coronavirus Disease 2019 (COVID-19) Patients: Novel Insights for Long COVID Metabolic Sequelae. J Pharmacol Exp Ther 2024; 389:34-39. [PMID: 38336381 PMCID: PMC10949163 DOI: 10.1124/jpet.122.001253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Emerging evidence indicates that the relationship between coronavirus disease 2019 (COVID-19) and diabetes is 2-fold: 1) it is known that the presence of diabetes and other metabolic alterations poses a considerably high risk to develop a severe COVID-19; 2) patients who survived a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have an increased risk of developing new-onset diabetes. However, the mechanisms underlying this association are mostly unknown, and there are no reliable biomarkers to predict the development of new-onset diabetes. In the present study, we demonstrate that a specific microRNA (miR-34a) contained in circulating extracellular vesicles released by endothelial cells reliably predicts the risk of developing new-onset diabetes in COVID-19. This association was independent of age, sex, body mass index (BMI), hypertension, dyslipidemia, smoking status, and D-dimer. SIGNIFICANCE STATEMENT: We demonstrate for the first time that a specific microRNA (miR-34a) contained in circulating extracellular vesicles released by endothelial cells is able to reliably predict the risk of developing diabetes after having contracted coronavirus disease 2019 (COVID-19). This association was independent of age, sex, body mass index (BMI), hypertension, dyslipidemia, smoking status, and D-dimer. Our findings are also relevant when considering the emerging importance of post-acute sequelae of COVID-19, with systemic manifestations observed even months after viral negativization (long COVID).
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Stanislovas S Jankauskas
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Maria Virginia Manzi
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Jessica Gambardella
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Antonietta Coppola
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Urna Kansakar
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Raffaele Izzo
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Giuseppe Fiorentino
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Angela Lombardi
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Fahimeh Varzideh
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Daniela Sorriento
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Bruno Trimarco
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| | - Gaetano Santulli
- Department of Medicine, Einstein-Sinai Diabetes Research Center, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Aging Research (P.M., S.S.J., J.G., U.K., A.L., F.V., G.S.) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (G.S.), Albert Einstein College of Medicine, New York, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education, "Federico II" University, Naples, Italy (M.V.M., J.G., R.I., D.S., B.T., G.S.); Clinica Montevergine, Mercogliano, Avellino (P.M.); and COVID-19 Division, A.O.R.N. Ospedali dei Colli, Naples, Italy (A.C., G.F.)
| |
Collapse
|
16
|
Valencia I, Lumpuy-Castillo J, Magalhaes G, Sánchez-Ferrer CF, Lorenzo Ó, Peiró C. Mechanisms of endothelial activation, hypercoagulation and thrombosis in COVID-19: a link with diabetes mellitus. Cardiovasc Diabetol 2024; 23:75. [PMID: 38378550 PMCID: PMC10880237 DOI: 10.1186/s12933-023-02097-8] [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: 10/18/2023] [Accepted: 12/14/2023] [Indexed: 02/22/2024] Open
Abstract
Early since the onset of the COVID-19 pandemic, the medical and scientific community were aware of extra respiratory actions of SARS-CoV-2 infection. Endothelitis, hypercoagulation, and hypofibrinolysis were identified in COVID-19 patients as subsequent responses of endothelial dysfunction. Activation of the endothelial barrier may increase the severity of the disease and contribute to long-COVID syndrome and post-COVID sequelae. Besides, it may cause alterations in primary, secondary, and tertiary hemostasis. Importantly, these responses have been highly decisive in the evolution of infected patients also diagnosed with diabetes mellitus (DM), who showed previous endothelial dysfunction. In this review, we provide an overview of the potential triggers of endothelial activation related to COVID-19 and COVID-19 under diabetic milieu. Several mechanisms are induced by both the viral particle itself and by the subsequent immune-defensive response (i.e., NF-κB/NLRP3 inflammasome pathway, vasoactive peptides, cytokine storm, NETosis, activation of the complement system). Alterations in coagulation mediators such as factor VIII, fibrin, tissue factor, the von Willebrand factor: ADAMST-13 ratio, and the kallikrein-kinin or plasminogen-plasmin systems have been reported. Moreover, an imbalance of thrombotic and thrombolytic (tPA, PAI-I, fibrinogen) factors favors hypercoagulation and hypofibrinolysis. In the context of DM, these mechanisms can be exacerbated leading to higher loss of hemostasis. However, a series of therapeutic strategies targeting the activated endothelium such as specific antibodies or inhibitors against thrombin, key cytokines, factor X, complement system, the kallikrein-kinin system or NETosis, might represent new opportunities to address this hypercoagulable state present in COVID-19 and DM. Antidiabetics may also ameliorate endothelial dysfunction, inflammation, and platelet aggregation. By improving the microvascular pathology in COVID-19 and post-COVID subjects, the associated comorbidities and the risk of mortality could be reduced.
Collapse
Affiliation(s)
- Inés Valencia
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, IIS Hospital Universitario de La Princesa, 28009, Madrid, Spain.
| | - Jairo Lumpuy-Castillo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040, Madrid, Spain
- Spanish Biomedical Research Centre On Diabetes and Associated Metabolic Disorders (CIBERDEM) Network, Madrid, Spain
| | - Giselle Magalhaes
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain
- Vascular Pharmacology and Metabolism (FARMAVASM), IdiPAZ, Madrid, Spain
| | - Óscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040, Madrid, Spain.
- Spanish Biomedical Research Centre On Diabetes and Associated Metabolic Disorders (CIBERDEM) Network, Madrid, Spain.
| | - Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029, Madrid, Spain.
- Vascular Pharmacology and Metabolism (FARMAVASM), IdiPAZ, Madrid, Spain.
| |
Collapse
|
17
|
Gauvin J, Huynh DN, Dubuc I, Lê C, Tugores R, Flamand N, Flamand L, Lubell WD, Ong H, Marleau S. Pharmacological targeting of the hyper-inflammatory response to SARS-CoV-2-infected K18-hACE2 mice using a cluster of differentiation 36 receptor modulator. Front Pharmacol 2024; 15:1303342. [PMID: 38384295 PMCID: PMC10879382 DOI: 10.3389/fphar.2024.1303342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
The scientific and medical community faced an unprecedented global health hazard that led to nearly 7 million deaths attributable to the rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In spite of the development of efficient vaccines against SARS-CoV-2, many people remain at risk of developing severe symptoms as the virus continues to spread without beneficial patient therapy. The hyper-inflammatory response to SARS-CoV-2 infection progressing to acute respiratory distress syndrome remains an unmet medical need for improving patient care. The viral infection stimulates alveolar macrophages to adopt an inflammatory phenotype regulated, at least in part, by the cluster of differentiation 36 receptor (CD36) to produce unrestrained inflammatory cytokine secretions. We suggest herein that the modulation of the macrophage response using the synthetic CD36 ligand hexarelin offers potential as therapy for halting respiratory failure in SARS-CoV-2-infected patients.
Collapse
Affiliation(s)
- Jade Gauvin
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - David N. Huynh
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Isabelle Dubuc
- Department of Microbiology, Infectious Diseases and and Immunology, Université Laval, Québec, QC, Canada
| | - Catherine Lê
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Rafaela Tugores
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Nicolas Flamand
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Louis Flamand
- Department of Microbiology, Infectious Diseases and and Immunology, Université Laval, Québec, QC, Canada
| | - William D. Lubell
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
18
|
Üstündağ H, Demir Ö, Huyut MT, Yüce N. Investigating the individual and combined effects of coenzyme Q10 and vitamin C on CLP-induced cardiac injury in rats. Sci Rep 2024; 14:3098. [PMID: 38326366 PMCID: PMC10850075 DOI: 10.1038/s41598-024-52932-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/25/2024] [Indexed: 02/09/2024] Open
Abstract
Sepsis-induced cardiac injury represents a major clinical challenge, amplifying the urgency for effective therapeutic interventions. This study aimed to delve into the individual and combined prophylactic effects of Vitamin C (Vit C) and Coenzyme Q10 (CoQ10) against inflammatory heart injury in a cecal ligation and puncture (CLP) induced polymicrobial sepsis rat model. Thirty adult female Sprague-Dawley rats were randomly divided into five groups: Control, CLP, Vitamin C, CoQ10, and Vit C + CoQ10, each consisting of six rats. Treatments were administered orally via gavage for 10 days prior to the operation. Eighteen hours post-sepsis induction, the animals were euthanized, and specimens were collected for analysis. The study examined variations in oxidative (TOS, OSI, MDA, MPO) and antioxidative markers (TAS, SOD, CAT, GSH), histopathological changes, inflammatory cytokine concentrations (TNF-α, IL-1β), nitric oxide (NO) dynamics, and cardiac indicators such as CK-MB. Impressively, the combined regimen markedly diminished oxidative stress, and antioxidative parameters reflected notable enhancements. Elevated NO levels, a central player in sepsis-driven inflammatory cascades, were effectively tempered by our intervention. Histological examinations corroborated the biochemical data, revealing diminished cardiac tissue damage in treated subjects. Furthermore, a marked suppression in pro-inflammatory cytokines was discerned, solidifying the therapeutic potential of our intervention. Interestingly, in certain evaluations, CoQ10 exhibited superior benefits over Vit C. Collectively, these findings underscore the potential therapeutic promise of Vit C and CoQ10 combination against septic cardiac injuries in rats.
Collapse
Affiliation(s)
- Hilal Üstündağ
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Türkiye.
| | - Özlem Demir
- Department of Histology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Mehmet Tahir Huyut
- Department of Biostatistics, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Neslihan Yüce
- Department of Biochemistry, Faculty of Medicine, Atatürk University, Erzurum, Türkiye
| |
Collapse
|
19
|
Perico L, Benigni A, Remuzzi G. SARS-CoV-2 and the spike protein in endotheliopathy. Trends Microbiol 2024; 32:53-67. [PMID: 37393180 PMCID: PMC10258582 DOI: 10.1016/j.tim.2023.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/03/2023]
Abstract
SARS-CoV-2, the causative agent of COVID-19, primarily affects the epithelial compartment in the upper and lower airways. There is evidence that the microvasculature in both the pulmonary and extrapulmonary systems is a major target of SARS-CoV-2. Consistent with this, vascular dysfunction and thrombosis are the most severe complications in COVID-19. The proinflammatory milieu triggered by the hyperactivation of the immune system by SARS-CoV-2 has been suggested to be the main trigger for endothelial dysfunction during COVID-19. More recently, a rapidly growing number of reports have indicated that SARS-CoV-2 can interact directly with endothelial cells through the spike protein, leading to multiple instances of endothelial dysfunction. Here, we describe all the available findings showing the direct effect of the SARS-CoV-2 spike protein on endothelial cells and offer mechanistic insights into the molecular basis of vascular dysfunction in severe COVID-19.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy.
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| |
Collapse
|
20
|
Luchian ML, Higny J, Benoit M, Robaye B, Berners Y, Henry JP, Colle B, Xhaët O, Blommaert D, Droogmans S, Motoc AI, Cosyns B, Gabriel L, Guedes A, Demeure F. Unmasking Pandemic Echoes: An In-Depth Review of Long COVID's Unabated Cardiovascular Consequences beyond 2020. Diagnostics (Basel) 2023; 13:3368. [PMID: 37958264 PMCID: PMC10647305 DOI: 10.3390/diagnostics13213368] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
At the beginning of 2020, coronavirus disease 2019 (COVID-19) emerged as a new pandemic, leading to a worldwide health crisis and overwhelming healthcare systems due to high numbers of hospital admissions, insufficient resources, and a lack of standardized therapeutic protocols. Multiple genetic variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been detected since its first public declaration in 2020, some of them being considered variants of concern (VOCs) corresponding to several pandemic waves. Nevertheless, a growing number of COVID-19 patients are continuously discharged from hospitals, remaining symptomatic even months after their first episode of COVID-19 infection. Long COVID-19 or 'post-acute COVID-19 syndrome' emerged as the new pandemic, being characterized by a high variability of clinical manifestations ranging from cardiorespiratory and neurological symptoms such as chest pain, exertional dyspnoea or cognitive disturbance to psychological disturbances, e.g., depression, anxiety or sleep disturbance with a crucial impact on patients' quality of life. Moreover, Long COVID is viewed as a new cardiovascular risk factor capable of modifying the trajectory of current and future cardiovascular diseases, altering the patients' prognosis. Therefore, in this review we address the current definitions of Long COVID and its pathophysiology, with a focus on cardiovascular manifestations. Furthermore, we aim to review the mechanisms of acute and chronic cardiac injury and the variety of cardiovascular sequelae observed in recovered COVID-19 patients, in addition to the potential role of Long COVID clinics in the medical management of this new condition. We will further address the role of future research for a better understanding of the actual impact of Long COVID and future therapeutic directions.
Collapse
Affiliation(s)
- Maria-Luiza Luchian
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Julien Higny
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Martin Benoit
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Benoit Robaye
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Yannick Berners
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Jean-Philippe Henry
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Benjamin Colle
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Olivier Xhaët
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Dominique Blommaert
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Steven Droogmans
- Department of Cardiology, Centrum voor Hart-en Vaatziekten, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Andreea Iulia Motoc
- Department of Cardiology, Centrum voor Hart-en Vaatziekten, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Bernard Cosyns
- Department of Cardiology, Centrum voor Hart-en Vaatziekten, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Laurence Gabriel
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Antoine Guedes
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| | - Fabian Demeure
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur Site Godinne, Av. Dr. G. Thérasse, 1, 5530 Yvoir, Belgium (A.G.); (F.D.)
| |
Collapse
|
21
|
Wang G, Lian H, Zhang H, Wang X. Microcirculation and Mitochondria: The Critical Unit. J Clin Med 2023; 12:6453. [PMID: 37892591 PMCID: PMC10607663 DOI: 10.3390/jcm12206453] [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: 08/23/2023] [Revised: 09/22/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Critical illness is often accompanied by a hemodynamic imbalance between macrocirculation and microcirculation, as well as mitochondrial dysfunction. Microcirculatory disorders lead to abnormalities in the supply of oxygen to tissue cells, while mitochondrial dysfunction leads to abnormal energy metabolism and impaired tissue oxygen utilization, making these conditions important pathogenic factors of critical illness. At the same time, there is a close relationship between the microcirculation and mitochondria. We introduce here the concept of a "critical unit", with two core components: microcirculation, which mainly comprises the microvascular network and endothelial cells, especially the endothelial glycocalyx; and mitochondria, which are mainly involved in energy metabolism but perform other non-negligible functions. This review also introduces several techniques and devices that can be utilized for the real-time synchronous monitoring of the microcirculation and mitochondria, and thus critical unit monitoring. Finally, we put forward the concepts and strategies of critical unit-guided treatment.
Collapse
Affiliation(s)
- Guangjian Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (G.W.); (H.Z.)
| | - Hui Lian
- Department of Health Care, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China;
| | - Hongmin Zhang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (G.W.); (H.Z.)
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (G.W.); (H.Z.)
| |
Collapse
|
22
|
Chen Z, Tang L, Luo L, Luo W, Li Y, Wang X, Huang L, Hu Y, Mei H. Enhancing the Treatment of Uncontrolled Inflammation through the Targeted Delivery of TPCA-1-Loaded Nanoparticles. Pharmaceutics 2023; 15:2435. [PMID: 37896195 PMCID: PMC10609852 DOI: 10.3390/pharmaceutics15102435] [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: 07/25/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Uncontrolled inflammation is a pathological state that underlies many diseases. Despite the development of numerous anti-inflammatory agents, the treatment of uncontrolled inflammation remains a challenging task. We developed a targeted delivery system for [5-(p-fluorophenyl)-2-ureido]thiophene-3-carboxamide (TPCA-1), a potent inhibitor of the NF-κB signaling pathway. The system comprises TPCA-1-loaded nanoparticles (NPs) functionalized with a monoclonal antibody (mAb) that specifically binds to the break point of the IgD6 region of the platelet/endothelial cell adhesion molecule-1 (PECAM-1) extracellular segment that is overexposed on the injured endothelium and activated macrophages during the pathogenesis of inflammation. In vitro binding and cellular uptake experiments revealed that the mAb modification on NPs could significantly enhance uptake by both Raw264.7 and HUVEC compared with unmodified NPs. In studies conducted at the cellular level focusing on anti-inflammatory and antioxidant effects, this formulation was found to effectively inhibit M1 polarization of macrophages, downregulate the secretion of pro-inflammatory cytokines, and reduce the production of reactive oxygen species (ROS) and nitric oxide (NO). In an animal model of vascular endothelial injury with acute inflammation, these NPs were capable of delivering TPCA-1 to inflammatory lesions in a targeted manner. Compared with the free agent-treated group, the NP-treated group exhibited reduced infiltration of inflammatory cells. In conclusion, our study demonstrates that this targeted delivery of TPCA-1-loaded NPs represents a promising strategy for improved mitigation of uncontrolled inflammation.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Lili Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Wenjing Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Yingying Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Xindi Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Linlin Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| |
Collapse
|
23
|
Kramer D, Hilton R, Roman J. Pulmonary fibrosis and COVID-19. Am J Med Sci 2023; 366:245-253. [PMID: 37481205 DOI: 10.1016/j.amjms.2023.07.006] [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: 04/08/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
The COVID-19 pandemic has caused the death of millions and many more have been infected worldwide. The causative virus, SARS-CoV-2, affects the lung where it elicits an aggressive inflammatory response leading to respiratory failure in severe cases. This infection has been linked to pulmonary fibrosis, a process characterized by fibroproliferation and the exaggerated deposition of collagen and other extracellular matrices. These events damage the lung architecture, especially its gas-exchanging units, leading to hypoxemic respiratory failure. The mechanisms by which the virus affects the lung remain incompletely understood, but it is postulated that after entering the airways, the virus binds to Angiotensin Converting Enzyme (ACE) receptors on the surface of epithelial cells, not only stimulating oxidative stress and inflammation, but also promoting the expression of soluble pro-fibrotic factors responsible for the accumulation of fibroblasts, their activation into myofibroblasts, and their unregulated expression of extracellular matrices. These events may trigger the rapid progression or exacerbation of underlying interstitial lung disorders or promote fibrosis in a previously healthy lung. Although the natural progression of such conditions cannot always be predicted, fibrosis may progress even after the virus has been eliminated or, in cases where it does not progress, may become irreversible, leading to long-standing symptoms like shortness of breath and exercise intolerance resulting from loss of lung function. Although COVID-19 related pulmonary fibrosis is not common, preventive measures like vaccination are encouraged, as they are expected to reduce infection or its severity, thereby decreasing the possibility of life-changing respiratory conditions such as pulmonary fibrosis.
Collapse
Affiliation(s)
- Daniel Kramer
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine; Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert Hilton
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine; Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jesse Roman
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine; Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
24
|
Greistorfer T, Jud P. Pathophysiological Aspects of COVID-19-Associated Vasculopathic Diseases. Thromb Haemost 2023; 123:931-944. [PMID: 37172941 DOI: 10.1055/s-0043-1768969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Since the beginning of coronavirus disease 2019 (COVID-19) pandemic, numerous data reported potential effects on the cardiovascular system due to infection by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), which may lead to COVID-19-associated vasculopathies during the acute phase and measurable vascular changes in the convalescent phase. Infection by SARS-CoV-2 seems to have specific direct and indirect effects on the endothelium, immune and coagulation systems thus promoting endothelial dysfunction, immunothrombosis, and formation of neutrophil extracellular traps although the exact mechanisms still need to be elucidated. This review represents a recent update of pathophysiological pathways of the respective three major mechanisms contributing to COVID-19 vasculopathies and vascular changes and includes clinical implications and significance of outcome data.
Collapse
Affiliation(s)
- Thiemo Greistorfer
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp Jud
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
25
|
Fields NJ, Palmer KR, Nisi A, Marshall SA. Preeclampsia to COVID-19: A journey towards improved placental and vascular function using sulforaphane. Placenta 2023; 141:84-93. [PMID: 37591715 DOI: 10.1016/j.placenta.2023.08.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023]
Abstract
Excess inflammation and oxidative stress are common themes in many pathologies of pregnancy including preeclampsia and more recently severe COVID-19. The risk of preeclampsia increases following maternal infection with COVID-19, potentially relating to significant overlap in pathophysiology with endothelial, vascular and immunological dysfunction common to both. Identifying a therapy which addresses these injurious processes and stabilises the endothelial and vascular maternal system would help address the significant global burden of maternal and neonatal morbidity and mortality they cause. Sulforaphane is a naturally occurring phytonutrient found most densely within cruciferous vegetables. It has anti-inflammatory, antioxidant and immune modulating properties via upregulation of phase-II detoxification enzymes. This review will cover the common pathways shared by COVID-19 and preeclampsia and offer a potential therapeutic target via nuclear factor erythroid 2-related factor upregulation in the form of sulforaphane.
Collapse
Affiliation(s)
- Neville J Fields
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, 27-31 Wright Street, Clayton, Victoria, Australia; Monash Health, Monash Medical Centre, 246 Clayton Road, Clayton, Victoria, Australia.
| | - Kirsten R Palmer
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, 27-31 Wright Street, Clayton, Victoria, Australia; Monash Health, Monash Medical Centre, 246 Clayton Road, Clayton, Victoria, Australia
| | - Anthony Nisi
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, 27-31 Wright Street, Clayton, Victoria, Australia
| | - Sarah A Marshall
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, 27-31 Wright Street, Clayton, Victoria, Australia
| |
Collapse
|
26
|
Tucci AR, da Rosa RM, Rosa AS, Augusto Chaves O, Ferreira VNS, Oliveira TKF, Coutinho Souza DD, Borba NRR, Dornelles L, Rocha NS, Mayer JCP, da Rocha JBT, Rodrigues OED, Miranda MD. Antiviral Effect of 5'-Arylchalcogeno-3-aminothymidine Derivatives in SARS-CoV-2 Infection. Molecules 2023; 28:6696. [PMID: 37764472 PMCID: PMC10537738 DOI: 10.3390/molecules28186696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/04/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The understanding that zidovudine (ZDV or azidothymidine, AZT) inhibits the RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 and that chalcogen atoms can increase the bioactivity and reduce the toxicity of AZT has directed our search for the discovery of novel potential anti-coronavirus compounds. Here, the antiviral activity of selenium and tellurium containing AZT derivatives in human type II pneumocytes cell model (Calu-3) and monkey kidney cells (Vero E6) infected with SARS-CoV-2, and their toxic effects on these cells, was evaluated. Cell viability analysis revealed that organoselenium (R3a-R3e) showed lower cytotoxicity than organotellurium (R3f, R3n-R3q), with CC50 ≥ 100 µM. The R3b and R3e were particularly noteworthy for inhibiting viral replication in both cell models and showed better selectivity index. In Vero E6, the EC50 values for R3b and R3e were 2.97 ± 0.62 µM and 1.99 ± 0.42 µM, respectively, while in Calu-3, concentrations of 3.82 ± 1.42 µM and 1.92 ± 0.43 µM (24 h treatment) and 1.33 ± 0.35 µM and 2.31 ± 0.54 µM (48 h) were observed, respectively. The molecular docking calculations were carried out to main protease (Mpro), papain-like protease (PLpro), and RdRp following non-competitive, competitive, and allosteric inhibitory approaches. The in silico results suggested that the organoselenium is a potential non-competitive inhibitor of RdRp, interacting in the allosteric cavity located in the palm region. Overall, the cell-based results indicated that the chalcogen-zidovudine derivatives were more potent than AZT in inhibiting SARS-CoV-2 replication and that the compounds R3b and R3e play an important inhibitory role, expanding the knowledge about the promising therapeutic capacity of organoselenium against COVID-19.
Collapse
Affiliation(s)
- Amanda Resende Tucci
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Raquel Mello da Rosa
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Alice Santos Rosa
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Otávio Augusto Chaves
- CQC-IMS, Departamento de Química, Universidade de Coimbra, Rua Larga, 3004-535 Coimbra, Portugal
- Laboratório de Imunofarmacologia, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias (CPIV), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-900, RJ, Brazil
| | - Vivian Neuza Santos Ferreira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
| | - Thamara Kelcya Fonseca Oliveira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Daniel Dias Coutinho Souza
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Nathalia Roberto Resende Borba
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
| | - Luciano Dornelles
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Nayra Salazar Rocha
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - João Candido Pilar Mayer
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - João B. Teixeira da Rocha
- Programa de Pós-Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil;
| | - Oscar Endrigo D. Rodrigues
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Milene Dias Miranda
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| |
Collapse
|
27
|
Vrbic M, Milinkovic A. Two vicious circles associated with the aging of the immune system in the development of severe forms of COVID-19. FRONTIERS IN AGING 2023; 4:1260053. [PMID: 37780864 PMCID: PMC10537960 DOI: 10.3389/fragi.2023.1260053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023]
Abstract
Background: The immune-inflammatory response is the basis of the pathophysiology of SARS-Cov-2 infection. In severe cases of COVID-19 uncontrolled systemic inflammatory response causes multiorgan dysfunction (MODS), as the most common immediate cause of death. Unfavorable outcome of the COVID-19 most often occurs in elderly patients. The aim of the study was to establish parameters with prognostic significance in severe cases of COVID-19 according to life years, laboratory markers of sepsis and MODS, as well as the number of peripheral CD4+ and CD8+T lymphocytes in 20 consecutively selected critically ill patients. Results: Eleven subjects were male, 9 female, mean age 73.45 ± 11.59, among which the oldest patient was 94 and the youngest 43 years. All the patients met the sepsis and MODS criteria. Increased age and low CD4+ and CD8+T cell counts were identified as independent predictors of death. Only the two youngest patients (43 and 50 years old) survived 28 days, and they are the only ones with a CD4 lymphocyte count above 500 cells/mm3. Conclusion: Senescence of the immune system is mostly characterized by reduced regenerative capacity of adaptive immunity with diminished ability to respond to new antigens and a manifested proinflammatory phenotype. Additional reduction of protective capacity by further deterioration of T cell quantity and quality due to sepsis itself and mutual interaction of senescent T cells and vascular endothelial cells in the induction of cytokine storm represent two complementary vicious cycles in the development of sepsis-related multiorgan dysfunction.
Collapse
Affiliation(s)
| | - Ana Milinkovic
- Chelsea and Westminster Foundation Trust and Imperial College London, London, United Kingdom
| |
Collapse
|
28
|
Avotins L, Kroica J, Petersons A, Zentina D, Kravale Z, Saulite A, Racenis K. eGFR cystatinC/eGFR creatinine ratio < 0.6 in patients with SARS-CoV-2 pneumonia: a prospective cohort study. BMC Nephrol 2023; 24:269. [PMID: 37704948 PMCID: PMC10500727 DOI: 10.1186/s12882-023-03315-x] [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: 04/15/2023] [Accepted: 08/29/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Shrunken Pore Syndrome (SPS), defined as a reduced ratio between two estimated filtration rates (based on cystatin C and creatinine) is an increasingly recognized risk factor for long-term mortality. Although some patients with other conditions might be erroneously identified as SPS. Our aim was to bring the focus on possible pathophysiologic mechanisms influencing the ratio in the setting of SARS-CoV-2 pneumonia and acute kidney injury. METHODS A single-centered prospective cohort study was conducted to investigate biomarkers in symptomatic COVID-19 pneumonia patients admitted to a hospital in Latvia. Nineteen biomarkers were measured in blood and three in urine samples. Associations were sought between these biomarkers, chronic diseases and the estimated GFRcystatinC/eGFRcreatinine ratio < 0.6, mortality rates, and acute kidney injury development. Data analysis was performed using SPSS Statistics, with significance set at p < 0.05. RESULTS We included 59 patients (average age 65.5 years, 45.8% female) admitted with COVID-19. Acute kidney injury occurred in 27.1%, and 25.4% died. Ratio < 0.6 was seen in 38.6%, associated with female sex, diabetes, hypothyroidism, and higher age. Ratio < 0.6 group had mortality notably higher - 40.9% vs. 16.2% and more cases of acute kidney injury (40.9% vs. 18.9%). Cystatin C showed strong associations with the ratio < 0.6 compared to creatinine. Urea levels and urea/creatinine ratio were higher in the ratio < 0.6 group. After excluding acute kidney injury patients, ratio < 0.6 remained associated with higher cystatin C and urea levels. Other biomarkers linked to a kidney injury as NGAL, and proteinuria did not differ. CONCLUSION We prove that reduced ratio is common in hospitalized patients with SARS-CoV-2 pneumonia and is associated with increased mortality during hospitalization. Factors that influence this ratio are complex and, in addition to the possible shrinkage of pores, other conditions such as thickening of glomerular basal membrane, comorbidities, prerenal kidney failure and others may play an important role and should be addressed when diagnosing SPS. We highlight the need for additional diagnostic criteria for SPS and larger studies to better understand its implications in acute COVID-19 settings.
Collapse
Affiliation(s)
- Lauris Avotins
- Rīga Stradiņš University, Riga, Latvia.
- Pauls Stradins Clinical University Hospital, Riga, Latvia.
| | | | - Aivars Petersons
- Rīga Stradiņš University, Riga, Latvia
- Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Dace Zentina
- Rīga Stradiņš University, Riga, Latvia
- Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Zaiga Kravale
- Rīga Stradiņš University, Riga, Latvia
- Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Anna Saulite
- Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Karlis Racenis
- Rīga Stradiņš University, Riga, Latvia
- Pauls Stradins Clinical University Hospital, Riga, Latvia
| |
Collapse
|
29
|
Aedo-Sánchez C, Gutiérrez G, Aguilar-Vidal E. COVID-19 and Vestibular Symptoms and Assessment: A Review. Audiol Neurootol 2023; 29:81-87. [PMID: 37703853 PMCID: PMC11412481 DOI: 10.1159/000533448] [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: 08/31/2022] [Accepted: 08/03/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND The current pandemic of COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in significant morbidity and mortality primarily associated with respiratory failure. However, it has also been reported that COVID-19 can evolve into a nervous system infection. The direct and indirect mechanisms of damage associated with SARS-CoV-2 neuropathogenesis could affect our sensory functionality, including hearing and balance. SUMMARY In order to investigate a possible association between SARS-CoV-2 viral infection and possible damage to the vestibular system, this review describes the main findings related to diagnosing and evaluating otoneurological pathologies. KEY MESSAGES The clinical evidence shows that SARS-CoV-2 causes acute damage to the vestibular system that would not leave significant sequelae. Recovery is similar to vestibular pathologies such as vestibular neuronitis and benign paroxysmal positional vertigo. Further basic science, clinical, and translational research is needed to verify and understand the short- and long-term effects of COVID-19 on vestibular function.
Collapse
Affiliation(s)
- Cristian Aedo-Sánchez
- Departamento Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gabriela Gutiérrez
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Enzo Aguilar-Vidal
- Departamento Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
30
|
Mohseni Afshar Z, Tavakoli Pirzaman A, Hosseinzadeh R, Babazadeh A, Taghizadeh Moghadam MA, Miri SR, Sio TT, Sullman MJM, Barary M, Ebrahimpour S. Anticoagulant therapy in COVID-19: A narrative review. Clin Transl Sci 2023; 16:1510-1525. [PMID: 37326220 PMCID: PMC10499427 DOI: 10.1111/cts.13569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/17/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can manifest itself in several ways, including coagulopathy and thrombosis. These complications can be the first and sometimes only manifestations of SARS-CoV-2 infection and can occur early or late in the course of the disease. However, these symptoms are more prevalent in hospitalized patients with venous thromboembolism, particularly those admitted to intensive care units. Moreover, various forms of arterial and venous thrombosis, or micro- or macro-vasculature embolisms, have been reported during the current pandemic. They have led to harmful consequences, such as neurological and cardiac events, nearly all resulting from the hypercoagulable state caused by this viral infection. The severe hypercoagulability observed in patients with COVID-19 accounts for most cases of the disease that become critical. Therefore, anticoagulants seem to be one of the most vital therapeutics for treating this potentially life-threatening condition. In the current paper, we present a thorough review of the pathophysiology of COVID-19-induced hypercoagulable state and the use of anticoagulants to treat SARS-CoV-2 infections in different patient groups, as well as their pros and cons.
Collapse
Affiliation(s)
- Zeinab Mohseni Afshar
- Clinical Research Development Center, Imam Reza HospitalKermanshah University of Medical SciencesKermanshahIran
| | | | | | - Arefeh Babazadeh
- Infectious Diseases and Tropical Medicine Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| | | | - Seyed Rouhollah Miri
- Cancer Research CenterCancer Institute of Iran, Tehran University of Medical ScienceTehranIran
| | - Terence T. Sio
- Department of Radiation OncologyMayo ClinicPhoenixArizonaUSA
| | - Mark J. M. Sullman
- Department of Social SciencesUniversity of NicosiaNicosiaCyprus
- Department of Life and Health SciencesUniversity of NicosiaNicosiaCyprus
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and ManagementShahid Beheshti University of Medical SciencesTehranIran
| | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| |
Collapse
|
31
|
Vishwakarma N, Goud RB, Tirupattur MP, Katwa LC. The Eye of the Storm: Investigating the Long-Term Cardiovascular Effects of COVID-19 and Variants. Cells 2023; 12:2154. [PMID: 37681886 PMCID: PMC10486388 DOI: 10.3390/cells12172154] [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: 07/26/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
COVID-19 had stormed through the world in early March of 2019, and on 5 May 2023, SARS-CoV-2 was officially declared to no longer be a global health emergency. The rise of new COVID-19 variants XBB.1.5 and XBB.1.16, a product of recombinant variants and sub-strains, has fueled a need for continued surveillance of the pandemic as they have been deemed increasingly infectious. Regardless of the severity of the variant, this has caused an increase in hospitalizations, a strain in resources, and a rise of concern for public health. In addition, there is a growing population of patients experiencing cardiovascular complications as a result of post-acute sequelae of COVID-19. This review aims to focus on what was known about SARS-CoV-2 and its past variants (Alpha, Delta, Omicron) and how the knowledge has grown today with new emerging variants, with an emphasis on cardiovascular complexities. We focus on the possible mechanisms that cause the observations of chronic cardiac conditions seen even after patients have recovered from the infection. Further understanding of these mechanisms will help to close the gap in knowledge on post-acute sequelae of COVID-19 and the differences between the effects of variants.
Collapse
Affiliation(s)
| | | | | | - Laxmansa C. Katwa
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA; (N.V.); (R.B.G.); (M.P.T.)
| |
Collapse
|
32
|
Ghondaghsaz E, Khalaji A, Norouzi M, Fraser DD, Alilou S, Behnoush AH. The utility of syndecan-1 circulating levels as a biomarker in patients with previous or active COVID-19: a systematic review and meta-analysis. BMC Infect Dis 2023; 23:510. [PMID: 37542221 PMCID: PMC10401738 DOI: 10.1186/s12879-023-08473-9] [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: 05/10/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND With the emergence of coronavirus disease of 2019 (COVID-19), several blood biomarkers have been identified, including the endothelial biomarker syndecan-1, a surface proteoglycan. In the current systematic review and meta-analysis, we aimed to assess the diagnostic and prognostic role of syndecan-1 in COVID-19. METHODS PubMed, Embase, Scopus, and Web of Science, as international databases, were searched for relevant studies measuring blood syndecan-1 levels in COVID-19 patients, COVID-19 convalescents, and healthy control subjects, in patients with different COVID-19 severities and/or in COVID-19 patients with poor outcomes. Random-effect meta-analysis was performed using STATA to calculate the standardized mean difference (SMD) and 95% confidence interval (CI) for the comparison between COVID-19 patients and healthy control subjects or COVID-19 convalescents and controls. RESULTS After screening by title/abstract and full text, 17 studies were included in the final review. Meta-analysis of syndecan-1 levels in COVID-19 compared with healthy control subjects revealed that patients with COVID-19 had significantly higher syndecan-1 levels (SMD 1.53, 95% CI 0.66 to 2.41, P < 0.01). In contrast, COVID-19 convalescent patients did not show significant difference with non-convalescents (SMD 0.08, 95% CI -0.63 to 0.78, P = 0.83). Regarding disease severity, two studies reported that more severe forms of the disease were associated with increased syndecan-1 levels. Moreover, patients who died from COVID-19 had higher syndecan-1 levels compared with survivors (SMD 1.22, 95% CI 0.10 to 2.33, P = 0.03). CONCLUSION Circulating syndecan-1 level can be used as a biomarker of endothelial dysfunction in COVID-19, as it was increased in COVID-19 patients and was higher in more severe instances of the disease. Further larger studies are needed to confirm these findings and further enlighten the role of syndecan-1 in clinical settings.
Collapse
Affiliation(s)
- Elina Ghondaghsaz
- Undergraduate Program in Neuroscience, University of British Columbia, Vancouver, BC Canada
| | - Amirmohammad Khalaji
- School of Medicine, Tehran University of Medical Sciences, Poursina St., Keshavarz Blvd, Tehran, 1417613151 Iran
| | - Mitra Norouzi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Douglas D. Fraser
- Children’s Health Research Institute, London, ON Canada
- Lawson Health Research Institute, London, ON Canada
- Department of Pediatrics, Western University, London, ON Canada
- Department of Physiology & Pharmacology, Western University, London, ON Canada
- Department of Clinical Neurological Sciences, Western University, London, ON Canada
| | - Sanam Alilou
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Behnoush
- School of Medicine, Tehran University of Medical Sciences, Poursina St., Keshavarz Blvd, Tehran, 1417613151 Iran
| |
Collapse
|
33
|
Owens CD, Bonin Pinto C, Mukli P, Szarvas Z, Peterfi A, Detwiler S, Olay L, Olson AL, Li G, Galvan V, Kirkpatrick AC, Balasubramanian P, Tarantini S, Csiszar A, Ungvari Z, Prodan CI, Yabluchanskiy A. Vascular mechanisms leading to progression of mild cognitive impairment to dementia after COVID-19: Protocol and methodology of a prospective longitudinal observational study. PLoS One 2023; 18:e0289508. [PMID: 37535668 PMCID: PMC10399897 DOI: 10.1371/journal.pone.0289508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023] Open
Abstract
INTRODUCTION Mild cognitive impairment (MCI) is a prodromal stage to dementia, affecting up to 20% of the aging population worldwide. Patients with MCI have an annual conversion rate to dementia of 15-20%. Thus, conditions that increase the conversion from MCI to dementia are of the utmost public health concern. The COVID-19 pandemic poses a significant impact on our aging population with cognitive decline as one of the leading complications following recovery from acute infection. Recent findings suggest that COVID-19 increases the conversion rate from MCI to dementia in older adults. Hence, we aim to uncover a mechanism for COVID-19 induced cognitive impairment and progression to dementia to pave the way for future therapeutic targets that may mitigate COVID-19 induced cognitive decline. METHODOLOGY A prospective longitudinal study is conducted at the University of Oklahoma Health Sciences Center. Patients are screened in the Department of Neurology and must have a formal diagnosis of MCI, and MRI imaging prior to study enrollment. Patients who meet the inclusion criteria are enrolled and followed-up at 18-months after their first visit. Visit one and 18-month follow-up will include an integrated and cohesive battery of vascular and cognitive measurements, including peripheral endothelial function (flow-mediated dilation, laser speckle contrast imaging), retinal and cerebrovascular hemodynamics (dynamic vessel retinal analysis, functional near-infrared spectroscopy), and fluid and crystalized intelligence (NIH-Toolbox, n-back). Multiple logistic regression will be used for primary longitudinal data analysis to determine whether COVID-19 related impairment in neurovascular coupling and increases in white matter hyperintensity burden contribute to progression to dementia.
Collapse
Affiliation(s)
- Cameron D. Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Camila Bonin Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Lauren Olay
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Ann L. Olson
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Guangpu Li
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Veronica Galvan
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Veterans Affairs Medical Center, Oklahoma City, OK, United States of America
| | - Angelia C. Kirkpatrick
- Veterans Affairs Medical Center, Oklahoma City, OK, United States of America
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Priya Balasubramanian
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Calin I. Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, United States of America
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| |
Collapse
|
34
|
Liu TH, Chuang MH, Wu JY, Huang PY, Tsai YW, Hsu WH, Lai CC. Effectiveness of oral antiviral agents on long-term cardiovascular risk in nonhospitalized patients with COVID-19: A multicenter matched cohort study. J Med Virol 2023; 95:e28992. [PMID: 37522355 DOI: 10.1002/jmv.28992] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/22/2023] [Accepted: 07/15/2023] [Indexed: 08/01/2023]
Abstract
Although a novel oral antiviral agent can improve short-term COVID-19 outcomes, its effects on the long-term outcomes, namely the risk of major adverse cardiovascular events (MACEs), remains unknown. This retrospective cohort study used the TriNetX research network to identify nonhospitalized adult patients with COVID-19 between March 1, 2020, and January 1, 2022. A propensity score matching method was used to form two matched cohorts with and without receiving nirmatrelvir-ritonavir (NMV-r) or molnupiravir. The primary outcome was the incidence of MACEs within a 30-day to 1-year period following a diagnosis of COVID-19. Two cohorts of each 80 888 patients with balanced baseline characteristics were formed using propensity score matching. During the follow-up period, 976 patients in the study group and 1609 patients in the control group developed MACE. Overall, the study group had a significantly lower risk of MACE than the control group (hazard ratio [HR], 0.683; 95% confidence interval: 0.630-0.739). The significantly lower HRs of overall MACEs were consistently observed in most subgroup analyses (age: >41-≤64 years: 0.60 [0.52-0.89]; age: ≥65 years: 0.68 [0.62-0.76]; women: 0.63 [0.57-0.71]; men: 0.62 [0.55-0.70]; vaccinated: 0.74 [0.63-0.88]; unvaccinated: 0.66 [0.60-0.73]; NMV-r; 0.65 [0.59-0.71]; and molnupiravir: 0.75 [0.61-0.92]). In conclusion, novel oral antiviral agents, namely NMV-r and molnupiravir, were effective in reducing long-term MACEs among nonhospitalized patients with COVID-19, particularly when treated with NMV-r or in patients aged ≥40 years. These findings suggest the potential role of novel antiviral agents as a preventive measure to reduce further adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Ting-Hui Liu
- Department of Psychiatry, Chi Mei Medical Center, Tainan, Taiwan
| | - Min-Hsiang Chuang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Jheng-Yan Wu
- Department of Nutrition, Chi Mei Medical Center, Tainan, Taiwan
| | - Po-Yu Huang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Ya-Wen Tsai
- Center of Integrative Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - Wan-Hsuan Hsu
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chih-Cheng Lai
- Department of Internal Medicine, Chi Mei Medical Center, Division of Hospital Medicine, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| |
Collapse
|
35
|
Bele A, Wagh V, Munjewar PK. A Comprehensive Review on Cardiovascular Complications of COVID-19: Unraveling the Link to Bacterial Endocarditis. Cureus 2023; 15:e44019. [PMID: 37746510 PMCID: PMC10517725 DOI: 10.7759/cureus.44019] [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: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
The global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has ushered in a new era of understanding the multifaceted nature of infectious diseases. Beyond its well-documented respiratory impact, COVID-19 has unveiled intricate interactions with the cardiovascular system, with potential implications that extend to bacterial endocarditis. This review explores the complex interplay between COVID-19 and bacterial endocarditis, elucidating shared risk factors, theoretical mechanisms, and clinical implications. We examine the diverse cardiovascular manifestations of COVID-19, ranging from myocarditis and thromboembolic events to arrhythmias, and delve into the pathogenesis, clinical features, and diagnostic challenges of bacterial endocarditis. By analyzing potential connections, such as viral-induced endothelial disruption and immune modulation, we shed light on the plausible relationship between COVID-19 and bacterial endocarditis. Our synthesis highlights the significance of accurate diagnosis, optimal management, and interdisciplinary collaboration in addressing the challenges posed by these intricate interactions. In addition, we underscore the importance of future research, emphasizing prospective studies on bacterial endocarditis incidence and investigations into the long-term cardiovascular effects of COVID-19. As the boundaries of infectious diseases and cardiovascular complications converge, this review calls for continued research, vigilance, and coordinated efforts to enhance patient care and public health strategies in a rapidly evolving landscape.
Collapse
Affiliation(s)
- Anurag Bele
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Vasant Wagh
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Pratiksha K Munjewar
- Medical Surgical Nursing, Smt. Radhikabai Meghe Memorial College of Nursing, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
36
|
Bykov VV, Motov VS, Bykova AV, Khazanov VA, Vengerovskii AI, Udut VV. Pharmacological Effects of a New Indolinone Derivative in Experimental Pneumonitis in Rats. Bull Exp Biol Med 2023; 175:459-462. [PMID: 37770782 DOI: 10.1007/s10517-023-05886-z] [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: 01/27/2023] [Indexed: 09/30/2023]
Abstract
We studied the effect of a new indolinone derivative GRS on animal survival and on functioning and histological structure of the lungs in rats with experimental pneumonitis. The rats of experimental groups were intratracheally administered 0.5% aqueous solution of carrageenan under intramuscular anesthesia. Compound GRS (10 mg/kg; a suspension in 0.5% aqueous solution of carboxymethyl cellulose) was orally administered for 4 days starting from the day of carrageenan administration; another rat group received the aqueous solution of carboxymethyl cellulose alone. Control rats received intratracheally isotonic solution of sodium chloride. Animal mortality was registered over 5 days; on day 5, the respiratory parameters were measured, the lungs were weighed, pulmonary edema was evaluated, and histological structure of the lungs was studied. GRS compound improved survival of animals with modeled pneumonitis, restored the respiratory parameters to the level of control animals, and reduced pulmonary edema by 35% and the severity of histological damage score in the lungs by 17% (p<0.05).
Collapse
Affiliation(s)
- V V Bykov
- LLC Innovative Pharmacology Research, Tomsk, Russia.
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - V S Motov
- LLC Innovative Pharmacology Research, Tomsk, Russia
| | - A V Bykova
- LLC Innovative Pharmacology Research, Tomsk, Russia
| | - V A Khazanov
- LLC Innovative Pharmacology Research, Tomsk, Russia
| | - A I Vengerovskii
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - V V Udut
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
37
|
Hwang YM, Piekos S, Sorensen T, Hood L, Hadlock J. Adoption of a National Prophylactic Anticoagulation Guideline for Hospitalized Pregnant Women With COVID-19: Retrospective Cohort Study. JMIR Public Health Surveill 2023; 9:e45586. [PMID: 37311123 PMCID: PMC10389076 DOI: 10.2196/45586] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/05/2023] [Accepted: 06/13/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Both COVID-19 and pregnancy are associated with hypercoagulability. Due to the increased risk for thrombosis, the United States National Institute of Health's recommendation for prophylactic anticoagulant use for pregnant patients has expanded from patients hospitalized for severe COVID-19 manifestation to all patients hospitalized for the manifestation of COVID-19 (no guideline: before December 26, 2020; first update: December 27, 2022; second update: February 24, 2022-present). However, no study has evaluated this recommendation. OBJECTIVE The objective of this study was to characterize prophylactic anticoagulant use among hospitalized pregnant people with COVID-19 from March 20, 2020, to October 19, 2022. METHODS This was a retrospective cohort study in large US health care systems across 7 states. The cohort of interest was pregnant patients who were hospitalized with COVID-19, without previous coagulopathy or contraindication to anticoagulants (n=2767). The treatment group consisted of patients prescribed prophylactic dose anticoagulation between 2 days before and 14 days after COVID-19 treatment onset (n=191). The control group was patients with no anticoagulant exposure between 14 days before and 60 days after COVID-19 treatment onset (n=2534). We ascertained the use of prophylactic anticoagulants with attention to the updates in guidelines and emerging SARS-CoV-2 variants. We propensity score matched the treatment and control group 1:1 on the most important features contributing to the prophylactic anticoagulant administration status classification. Outcome measures included coagulopathy, bleeding, COVID-19-related complications, and maternal-fetal health outcomes. Additionally, the inpatient anticoagulant administration rate was validated in a nationwide population from Truveta, a collective of 700 hospitals across the United States. RESULTS The overall administration rate of prophylactic anticoagulants was 7% (191/2725). It was lowest after the second guideline update (no guideline: 27/262, 10%; first update: 145/1663, 8.72%; second update: 19/811, 2.3%; P<.001) and during the omicron-dominant period (Wild type: 45/549, 8.2%; Alpha: 18/129, 14%; Delta: 81/507, 16%; and Omicron: 47/1551, 3%; P<.001). Models developed on retrospective data showed that the variable most associated with the administration of inpatient prophylactic anticoagulant was comorbidities prior to SARS-CoV-2 infection. The patients who were administered prophylactic anticoagulant were also more likely to receive supplementary oxygen (57/191, 30% vs 9/188, 5%; P<.001). There was no statistical difference in a new diagnosis of coagulopathy, bleeding, or maternal-fetal health outcomes between those who received treatment and the matched control group. CONCLUSIONS Most hospitalized pregnant patients with COVID-19 did not receive prophylactic anticoagulants across health care systems as recommended by guidelines. Guideline-recommended treatment was administered more frequently to patients with greater COVID-19 illness severity. Given the low rate of administration and differences between treated and untreated cohorts, efficacy could not be assessed.
Collapse
Affiliation(s)
- Yeon-Mi Hwang
- Institute for Systems Biology, Seattle, WA, United States
- University of Washington, Seattle, WA, United States
| | | | - Tanya Sorensen
- University of Washington, Seattle, WA, United States
- Swedish Medical Center, Providence Swedish, Seattle, WA, United States
| | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, United States
| | - Jennifer Hadlock
- Institute for Systems Biology, Seattle, WA, United States
- University of Washington, Seattle, WA, United States
| |
Collapse
|
38
|
Perico L, Morigi M, Pezzotta A, Locatelli M, Imberti B, Corna D, Cerullo D, Benigni A, Remuzzi G. SARS-CoV-2 spike protein induces lung endothelial cell dysfunction and thrombo-inflammation depending on the C3a/C3a receptor signalling. Sci Rep 2023; 13:11392. [PMID: 37452090 PMCID: PMC10349115 DOI: 10.1038/s41598-023-38382-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
The spike protein of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) can interact with endothelial cells. However, no studies demonstrated the direct effect of the spike protein subunit 1 (S1) in inducing lung vascular damage and the potential mechanisms contributing to lung injury. Here, we found that S1 injection in mice transgenic for human angiotensin converting enzyme 2 (ACE2) induced early loss of lung endothelial thromboresistance at 3 days, as revealed by thrombomodulin loss and von Willebrand factor (vWF) increase. In parallel, vascular and epithelial C3 deposits and enhanced C3a receptor (C3aR) expression were observed. These changes preceded diffuse alveolar damage and lung vascular fibrin(ogen)/platelets aggregates at 7 days, as well as inflammatory cell recruitment and fibrosis. Treatment with C3aR antagonist (C3aRa) inhibited lung C3 accumulation and C3a/C3aR activation, limiting vascular thrombo-inflammation and fibrosis. Our study demonstrates that S1 triggers vascular dysfunction and activates complement system, instrumental to lung thrombo-inflammatory injury. By extension, our data indicate C3aRa as a valuable therapeutic strategy to limit S1-dependent lung pathology.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| | - Marina Morigi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Anna Pezzotta
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Monica Locatelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Barbara Imberti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Daniela Corna
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Domenico Cerullo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
39
|
Reiss AB, Greene C, Dayaramani C, Rauchman SH, Stecker MM, De Leon J, Pinkhasov A. Long COVID, the Brain, Nerves, and Cognitive Function. Neurol Int 2023; 15:821-841. [PMID: 37489358 PMCID: PMC10366776 DOI: 10.3390/neurolint15030052] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/23/2023] [Accepted: 06/28/2023] [Indexed: 07/26/2023] Open
Abstract
SARS-CoV-2, a single-stranded RNA coronavirus, causes an illness known as coronavirus disease 2019 (COVID-19). Long-term complications are an increasing issue in patients who have been infected with COVID-19 and may be a result of viral-associated systemic and central nervous system inflammation or may arise from a virus-induced hypercoagulable state. COVID-19 may incite changes in brain function with a wide range of lingering symptoms. Patients often experience fatigue and may note brain fog, sensorimotor symptoms, and sleep disturbances. Prolonged neurological and neuropsychiatric symptoms are prevalent and can interfere substantially in everyday life, leading to a massive public health concern. The mechanistic pathways by which SARS-CoV-2 infection causes neurological sequelae are an important subject of ongoing research. Inflammation- induced blood-brain barrier permeability or viral neuro-invasion and direct nerve damage may be involved. Though the mechanisms are uncertain, the resulting symptoms have been documented from numerous patient reports and studies. This review examines the constellation and spectrum of nervous system symptoms seen in long COVID and incorporates information on the prevalence of these symptoms, contributing factors, and typical course. Although treatment options are generally lacking, potential therapeutic approaches for alleviating symptoms and improving quality of life are explored.
Collapse
Affiliation(s)
- Allison B Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Long Island, NY 11501, USA
| | - Caitriona Greene
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Long Island, NY 11501, USA
| | - Christopher Dayaramani
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Long Island, NY 11501, USA
| | | | | | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Long Island, NY 11501, USA
| | - Aaron Pinkhasov
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Long Island, NY 11501, USA
| |
Collapse
|
40
|
Owens CD, Pinto CB, Detwiler S, Mukli P, Peterfi A, Szarvas Z, Hoffmeister JR, Galindo J, Noori J, Kirkpatrick AC, Dasari TW, James J, Tarantini S, Csiszar A, Ungvari Z, Prodan CI, Yabluchanskiy A. Cerebral small vessel disease pathology in COVID-19 patients: A systematic review. Ageing Res Rev 2023; 88:101962. [PMID: 37224885 PMCID: PMC10202464 DOI: 10.1016/j.arr.2023.101962] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Cerebral small vessel disease (CSVD) is the leading cause of vascular cognitive impairment and is associated with COVID-19. However, contributing factors that often accompany CSVD pathology in COVID-19 patients may influence the incidence of cerebrovascular complications. Thus, a mechanism linking COVID-19 and CSVD has yet to be uncovered and differentiated from age-related comorbidities (i.e., hypertension), and medical interventions during acute infection. We aimed to evaluate CSVD in acute and recovered COVID-19 patients and to differentiate COVID-19-related cerebrovascular pathology from the above-mentioned contributing factors by assessing the localization of microbleeds and ischemic lesions/infarctions in the cerebrum, cerebellum, and brainstem. A systematic search was performed in December 2022 on PubMed, Web of Science, and Embase using a pre-established search criterion related to history of, or active COVID-19 with CSVD pathology in adults. From a pool of 161 studies, 59 met eligibility criteria and were included. Microbleeds and ischemic lesions had a strong predilection for the corpus callosum and subcortical/deep white matter in COVID-19 patients, suggesting a distinct CSVD pathology. These findings have important implications for clinical practice and biomedical research as COVID-19 may independently, and through exacerbation of age-related mechanisms, contribute to increased incidence of CSVD.
Collapse
Affiliation(s)
- Cameron D Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Camila Bonin Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Jordan R Hoffmeister
- Department of Psychiatry and Behavioral Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Juliette Galindo
- Department of Psychiatry and Behavioral Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jila Noori
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Angelia C Kirkpatrick
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Tarun W Dasari
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Judith James
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA; Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
41
|
Escaffre O, Szaniszlo P, Törő G, Vilas CL, Servantes BJ, Lopez E, Juelich TL, Levine CB, McLellan SLF, Cardenas JC, Freiberg AN, Módis K. Hydrogen Sulfide Ameliorates SARS-CoV-2-Associated Lung Endothelial Barrier Disruption. Biomedicines 2023; 11:1790. [PMID: 37509430 PMCID: PMC10376201 DOI: 10.3390/biomedicines11071790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 07/30/2023] Open
Abstract
Recent studies have confirmed that lung microvascular endothelial injury plays a critical role in the pathophysiology of COVID-19. Our group and others have demonstrated the beneficial effects of H2S in several pathological processes and provided a rationale for considering the therapeutic implications of H2S in COVID-19 therapy. Here, we evaluated the effect of the slow-releasing H2S donor, GYY4137, on the barrier function of a lung endothelial cell monolayer in vitro, after challenging the cells with plasma samples from COVID-19 patients or inactivated SARS-CoV-2 virus. We also assessed how the cytokine/chemokine profile of patients' plasma, endothelial barrier permeability, and disease severity correlated with each other. Alterations in barrier permeability after treatments with patient plasma, inactivated virus, and GYY4137 were monitored and assessed by electrical impedance measurements in real time. We present evidence that GYY4137 treatment reduced endothelial barrier permeability after plasma challenge and completely reversed the endothelial barrier disruption caused by inactivated SARS-CoV-2 virus. We also showed that disease severity correlated with the cytokine/chemokine profile of the plasma but not with barrier permeability changes in our assay. Overall, these data demonstrate that treatment with H2S-releasing compounds has the potential to ameliorate SARS-CoV-2-associated lung endothelial barrier disruption.
Collapse
Affiliation(s)
- Olivier Escaffre
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy & Smith Foundation, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Peter Szaniszlo
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gabor Törő
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Caitlyn L. Vilas
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Brenna J. Servantes
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ernesto Lopez
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Terry L. Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Corri B. Levine
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Susan L. F. McLellan
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jessica C. Cardenas
- The Center for Translational Injury Research, Department of Surgery, UTHealth McGovern Medical School, Houston, TX 77030, USA
| | - Alexander N. Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy & Smith Foundation, University of Texas Medical Branch, Galveston, TX 77555, USA
- The Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Katalin Módis
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
42
|
Xuereb RA, Borg M, Vella K, Gatt A, Xuereb RG, Barbara C, Fava S, Magri CJ. LONG COVID SYNDROME: A CASE-CONTROL STUDY. Am J Med 2023:S0002-9343(23)00299-1. [PMID: 37169323 PMCID: PMC10168190 DOI: 10.1016/j.amjmed.2023.04.022] [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: 05/21/2022] [Revised: 03/11/2023] [Accepted: 04/06/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Acute coronavirus disease 2019 (COVID-19) causes various cardiovascular complications. However, it is unknown if there are cardiovascular sequelae in the medium and long-term. The aim of this study was dual. Firstly, we wanted to investigate symptomology and health-related quality of life (HRQoL) at medium-term follow-up (6 months post-COVID). Secondly, we wanted to assess whether history of COVID-19 and persistent shortness of breath at medium-term follow-up are associated with ongoing inflammation, endothelial dysfunction and cardiac injury. METHODS A case-control study was performed. Virologically proven COVID-19 cases and age- and gender-matched controls were interviewed to assess symptoms and HRQoL. Biochemical tests were also performed. RESULTS The study comprised 174 cases and 75 controls. The mean participants' age was 46.1±13.8 years. The median follow-up was of 173.5 days (IQR 129-193.25 days). There was no significant difference in the demographics between cases and controls. At follow-up, cases had a higher frequency of shortness of breath, fatigue, arthralgia, abnormal taste of food (p<0.001) and anosmia. Cases also exhibited worse scores in the general health and role physical domains of the Short Form Survey-36. hsCRP was significantly higher in cases and there was a positive correlation of hsCRP with time. Significant determinants of shortness of breath were age, female gender and white cell count, troponin I and lower haemoglobin levels at follow-up. CONCLUSION Post-COVID-19 patients have persistent symptomatology at medium-term follow-up. Higher hsCRP in cases and the positive association of hsCRP with time suggest ongoing systemic inflammation in patients persisting for months after COVID-19.
Collapse
Affiliation(s)
- Rachel-Anne Xuereb
- Mater Dei Hospital, Msida, Malta; University of Malta Medical School, Msida, Malta
| | | | | | - Alex Gatt
- Mater Dei Hospital, Msida, Malta; University of Malta Medical School, Msida, Malta
| | - Robert G Xuereb
- Mater Dei Hospital, Msida, Malta; University of Malta Medical School, Msida, Malta
| | - Christopher Barbara
- Mater Dei Hospital, Msida, Malta; University of Malta Medical School, Msida, Malta
| | - Stephen Fava
- Mater Dei Hospital, Msida, Malta; University of Malta Medical School, Msida, Malta.
| | - Caroline J Magri
- Mater Dei Hospital, Msida, Malta; University of Malta Medical School, Msida, Malta
| |
Collapse
|
43
|
Thakur A, Liang L, Banerjee S, Zhang K. Single-Cell Transcriptomics Reveals Evidence of Endothelial Dysfunction in the Brains of COVID-19 Patients with Implications for Glioblastoma Progression. Brain Sci 2023; 13:brainsci13050762. [PMID: 37239234 DOI: 10.3390/brainsci13050762] [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: 04/01/2023] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Endothelial dysfunction is implicated in various inflammatory diseases such as ischemic stroke, heart attack, organ failure, and COVID-19. Recent studies have shown that endothelial dysfunction in the brain is attributed to excessive inflammatory responses caused by the SARS-CoV-2 infection, leading to increased permeability of the blood-brain barrier and consequently neurological damage. Here, we aim to examine the single-cell transcriptomic landscape of endothelial dysfunction in COVID-19 and its implications for glioblastoma (GBM) progression. METHODS Single-cell transcriptome data GSE131928 and GSE159812 were obtained from the gene expression omnibus (GEO) to analyze the expression profiles of key players in innate immunity and inflammation between brain endothelial dysfunction caused by COVID-19 and GBM progression. RESULTS Single-cell transcriptomic analysis of the brain of COVID-19 patients revealed that endothelial cells had undergone significant transcriptomic changes, with several genes involved in immune responses and inflammation upregulated. Moreover, transcription factors were observed to modulate this inflammation, including interferon-regulated genes. CONCLUSIONS The results indicate a significant overlap between COVID-19 and GBM in the context of endothelial dysfunction, suggesting that there may be an endothelial dysfunction link connecting severe SARS-CoV-2 infection in the brain to GBM progression.
Collapse
Affiliation(s)
- Abhimanyu Thakur
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation-CAS Limited, Hong Kong 999077, China
| | - Lifan Liang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Sourav Banerjee
- Department of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Kui Zhang
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400716, China
- Cancer Centre, Medical Research Institute, Southwest University, Chongqing 400716, China
| |
Collapse
|
44
|
Pincemail J, Rousseau AF, Kaux JF, Cheramy-Bien JP, Bruyère C, Prick J, Stern D, Kaci MM, Maertens De Noordhout B, Albert A, Eubelen C, Goff CL, Misset B, Cavalier E, Charlier C, Meziane S. A Pilot Study on Oxidative Stress during the Recovery Phase in Critical COVID-19 Patients in a Rehabilitation Facility: Potential Utility of the PAOT ® Technology for Assessing Total Anti-Oxidative Capacity. Biomedicines 2023; 11:biomedicines11051308. [PMID: 37238982 DOI: 10.3390/biomedicines11051308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/03/2023] [Accepted: 03/21/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Oxidative stress (OS) could cause various COVID-19 complications. Recently, we have developed the Pouvoir AntiOxydant Total (PAOT®) technology for reflecting the total antioxidant capacity (TAC) of biological samples. We aimed to investigate systemic oxidative stress status (OSS) and to evaluate the utility of PAOT® for assessing TAC during the recovery phase in critical COVID-19 patients in a rehabilitation facility. MATERIALS AND METHODS In a total of 12 critical COVID-19 patients in rehabilitation, 19 plasma OSS biomarkers were measured: antioxidants, TAC, trace elements, oxidative damage to lipids, and inflammatory biomarkers. TAC level was measured in plasma, saliva, skin, and urine, using PAOT and expressed as PAOT-Plasma, -Saliva, -Skin, and -Urine scores, respectively. Plasma OSS biomarker levels were compared with levels from previous studies on hospitalized COVID-19 patients and with the reference population. Correlations between four PAOT scores and plasma OSS biomarker levels were analyzed. RESULTS During the recovery phase, plasma levels in antioxidants (γ-tocopherol, β-carotene, total glutathione, vitamin C and thiol proteins) were significantly lower than reference intervals, whereas total hydroperoxides and myeloperoxidase (a marker of inflammation) were significantly higher. Copper negatively correlated with total hydroperoxides (r = 0.95, p = 0.001). A similar, deeply modified OSS was already observed in COVID-19 patients hospitalized in an intensive care unit. TAC evaluated in saliva, urine, and skin correlated negatively with copper and with plasma total hydroperoxides. To conclude, the systemic OSS, determined using a large number of biomarkers, was always significantly increased in cured COVID-19 patients during their recovery phase. The less costly evaluation of TAC using an electrochemical method could potentially represent a good alternative to the individual analysis of biomarkers linked to pro-oxidants.
Collapse
Affiliation(s)
- Joël Pincemail
- Clinical Chemistry, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | | | - Jean-François Kaux
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Jean-Paul Cheramy-Bien
- Department of Cardiovascular Surgery, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Christine Bruyère
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Jeanine Prick
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - David Stern
- Veterinary Medicine Faculty, FARAH, University of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Mouna-Messaouda Kaci
- Research Department, Institut Européen des Antioxydants (IEA), Oxystress Technologies PAOTScan, 54500 Vandœuvre-lès-Nancy, France
| | - Benoît Maertens De Noordhout
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Adelin Albert
- Biostatistics Department, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Céline Eubelen
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Caroline Le Goff
- Clinical Chemistry, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Benoît Misset
- Intensive Care Department, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Etienne Cavalier
- Clinical Chemistry, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Corinne Charlier
- Toxicology Department, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Smail Meziane
- Research Department, Institut Européen des Antioxydants (IEA), Oxystress Technologies PAOTScan, 54500 Vandœuvre-lès-Nancy, France
| |
Collapse
|
45
|
Sarkar S, Karmakar S, Basu M, Ghosh P, Ghosh MK. Neurological damages in COVID-19 patients: Mechanisms and preventive interventions. MedComm (Beijing) 2023; 4:e247. [PMID: 37035134 PMCID: PMC10080216 DOI: 10.1002/mco2.247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/14/2023] [Accepted: 03/01/2023] [Indexed: 04/11/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus, causes coronavirus disease 2019 (COVID-19) which led to neurological damage and increased mortality worldwide in its second and third waves. It is associated with systemic inflammation, myocardial infarction, neurological illness including ischemic strokes (e.g., cardiac and cerebral ischemia), and even death through multi-organ failure. At the early stage, the virus infects the lung epithelial cells and is slowly transmitted to the other organs including the gastrointestinal tract, blood vessels, kidneys, heart, and brain. The neurological effect of the virus is mainly due to hypoxia-driven reactive oxygen species (ROS) and generated cytokine storm. Internalization of SARS-CoV-2 triggers ROS production and modulation of the immunological cascade which ultimately initiates the hypercoagulable state and vascular thrombosis. Suppression of immunological machinery and inhibition of ROS play an important role in neurological disturbances. So, COVID-19 associated damage to the central nervous system, patients need special care to prevent multi-organ failure at later stages of disease progression. Here in this review, we are selectively discussing these issues and possible antioxidant-based prevention therapies for COVID-19-associated neurological damage that leads to multi-organ failure.
Collapse
Affiliation(s)
- Sibani Sarkar
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| | - Subhajit Karmakar
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| | - Malini Basu
- Department of MicrobiologyDhruba Chand Halder College, University of CalcuttaDakshin BarasatWBIndia
| | - Pratyasha Ghosh
- Department of EconomicsBethune CollegeUniversity of CalcuttaKolkataIndia
| | - Mrinal K Ghosh
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| |
Collapse
|
46
|
Votto M, Castagnoli R, Marseglia GL, Licari A, Brambilla I. COVID-19 and autoimmune diseases: is there a connection? Curr Opin Allergy Clin Immunol 2023; 23:185-192. [PMID: 36728317 DOI: 10.1097/aci.0000000000000888] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE OF REVIEW This review summarizes current evidence on the potential link between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and autoimmunity. RECENT FINDINGS Several viral infections are potential triggers of reactive and autoimmune diseases by inducing type II and type IV hypersensitivity reactions. Recent evidence demonstrated that SARS-CoV-2 infection is not an exception, triggering the production of tissue-specific autoantibodies during the acute phase of coronavirus disease 2019 (COVID-19) and leading to autoimmune diseases development as long-term complication. The significant immune dysregulation with cytokine storm and organ damage observed in patients with severe to critical COVID-19 is considered the main mechanism explaining the high levels of autoantibodies, which are also implicated in disease severity and the need for an intensive care assessment. Multisystem inflammatory syndrome in children (MIS-C) is an immune-mediated disease where the recent viral infection leads to systemic inflammation, as already observed in other reactive and autoimmune diseases. SUMMARY Autoimmunity may be a complication of SAR-CoV-2 infection. Understanding the pathogenesis of autoimmune manifestations in COVID-19 might help prevent the incidence or exacerbation of autoimmune disorders and design better and more efficient treatment strategies in children and adult populations.
Collapse
Affiliation(s)
- Martina Votto
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia
| | - Riccardo Castagnoli
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gian Luigi Marseglia
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Amelia Licari
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Ilaria Brambilla
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
47
|
COVID-19 and Vasa vasorum: New Atherogenic Factor? A Case Report and Autopsy Findings. Diagnostics (Basel) 2023; 13:diagnostics13061097. [PMID: 36980405 PMCID: PMC10047382 DOI: 10.3390/diagnostics13061097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Patients with COVID-19 demonstrate higher rates of cardiovascular complications, including thromboses and thromboembolism. One may suppose that the action of SARS-CoV-2 transforms stable atherosclerotic plaques into unstable status. Cardiovascular complications in COVID-19 may be caused by progressive viral alteration of the blood vessels, including Vasa vasorum. A lethal case of ischemic brain disease caused by cerebral atherosclerosis and exacerbated by a stroke during COVID-19 infection is briefly described. The results of the autopsy showed perivascular lymphocytic infiltration and signs of Vasa vasorum vasculitis with thrombi of adventitial microvasculature. The data discussed in the article are interpreted in the context of the concept giving the important role in atherogenesis to Vasa vasorum.
Collapse
|
48
|
Septimiu-Radu S, Gadela T, Gabriela D, Oancea C, Rosca O, Lazureanu VE, Fericean RM, Bratosin F, Dumitrescu A, Stoicescu ER, Bagiu I, Murariu M, Mavrea A. A Systematic Review of Lung Autopsy Findings in Elderly Patients after SARS-CoV-2 Infection. J Clin Med 2023; 12:2070. [PMID: 36902856 PMCID: PMC10004532 DOI: 10.3390/jcm12052070] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/27/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023] Open
Abstract
Although COVID-19 may cause various and multiorgan diseases, few research studies have examined the postmortem pathological findings of SARS-CoV-2-infected individuals who died. Active autopsy results may be crucial for understanding how COVID-19 infection operates and preventing severe effects. In contrast to younger persons, however, the patient's age, lifestyle, and concomitant comorbidities might alter the morpho-pathological aspects of the damaged lungs. Through a systematic analysis of the available literature until December 2022, we aimed to provide a thorough picture of the histopathological characteristics of the lungs in patients older than 70 years who died of COVID-19. A thorough search was conducted on three electronic databases (PubMed, Scopus, and Web of Science), including 18 studies and a total of 478 autopsies performed. It was observed that the average age of patients was 75.6 years, of which 65.4% were men. COPD was identified in an average of 16.7% of all patients. Autopsy findings indicated significantly heavier lungs, with an average weight of the right lung of 1103 g, while the left lung mass had an average weight of 848 g. Diffuse alveolar damage was a main finding in 67.2% of all autopsies, while pulmonary edema had a prevalence of between 50% and 70%. Thrombosis was also a significant finding, while some studies described focal and extensive pulmonary infarctions in 72.7% of elderly patients. Pneumonia and bronchopneumonia were observed, with a prevalence ranging from 47.6% to 89.5%. Other important findings described in less detail comprise hyaline membranes, the proliferation of pneumocytes and fibroblasts, extensive suppurative bronchopneumonic infiltrates, intra-alveolar edema, thickened alveolar septa, desquamation of pneumocytes, alveolar infiltrates, multinucleated giant cells, and intranuclear inclusion bodies. These findings should be corroborated with children's and adults' autopsies. Postmortem examination as a technique for studying the microscopic and macroscopic features of the lungs might lead to a better knowledge of COVID-19 pathogenesis, diagnosis, and treatment, hence enhancing elderly patient care.
Collapse
Affiliation(s)
- Susa Septimiu-Radu
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Tejaswi Gadela
- School of General Medicine, Bhaskar Medical College, Amdapur Road 156-162, Hyderabad 500075, India
| | - Doros Gabriela
- Department of Pediatrics, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Cristian Oancea
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ovidiu Rosca
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Voichita Elena Lazureanu
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Roxana Manuela Fericean
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Felix Bratosin
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Andreea Dumitrescu
- Cardioprevent Foundation, Calea Dorobantilor 3, Timisoara 300134, Romania
| | - Emil Robert Stoicescu
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Department of Radiology and Medical Imaging, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Iulia Bagiu
- Department of Microbiology, Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Mircea Murariu
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Department of Pediatrics, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Adelina Mavrea
- Department of Internal Medicine I, Cardiology Clinic, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| |
Collapse
|
49
|
Reștea PA, Mureșan M, Voicu A, Jurca T, Pallag A, Marian E, Vicaș LG, Jeican II, Crivii CB. Antidiabetic Treatment before Hospitalization and Admission Parameters in Patients with Type 2 Diabetes, Obesity, and SARS-CoV-2 Viral Infection. J Pers Med 2023; 13:jpm13030392. [PMID: 36983573 PMCID: PMC10055907 DOI: 10.3390/jpm13030392] [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: 12/25/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND SARS-CoV-2 viral infection is a current and important topic for patients with comorbidities of type 2 diabetes and obesity, associated with increased risk of mortality and morbidity. This study aims to analyze, compare and describe admission parameters in patients with type 2 diabetes, obesity, and SARS-CoV-2 infection based on whether they received insulin therapy before hospital admission. METHODS Our study enrolled patients diagnosed with type 2 diabetes, obesity, and SARS-CoV-2 viral infection, 81 patients without insulin treatment before hospital admission, and 81 patients with insulin at "Gavril Curteanu" Municipal Clinical Hospital of Oradea, Romania, between August 2020 and March 2022. RT-PCR/rapid antigen tests were used for detecting SARS-CoV-2 viral infection. RESULTS The severe form of COVID-19 was found in 66% of all patients (65% in the group without insulin and 67% in the group with insulin). Oxygen saturation at the time of hospital admission was greater or equal to 90% in 62% of all patients. The most associated comorbidities we founded in this study were: hypertension in 75% of all patients (grade two hypertension 63% in the group without insulin and 64% in the group with insulin), ischemic heart disease in 35% of patients (25% in the group without insulin and 44% in the group with insulin, n = 0.008), heart failure in 9.3% of all patients (8.6% in the group without insulin and 10% in the group with insulin). CRP and procalcitonin are increased in both groups at hospital admission, with a slightly higher trend in the group with insulin therapy before hospital admission. We found that 56% of patients in the group with insulin treatment were with uncontrolled diabetes on admission. Only 10% of patients required a change in antidiabetic treatment with insulin therapy at discharge. In our study, 89% of all patients did not require short-term home oxygen therapy at discharge. CONCLUSIONS Antidiabetic therapy taken before hospital admission did not protect patients against cytokine storm in COVID-19, but is very important in the pathophysiological stage of comorbidities. Paraclinical parameters at hospitalization showed differences in correlation with oral antidiabetic treatment like metformin or insulin therapy. Changing the antidiabetic treatment for a small percentage of patients in the group who had not been receiving insulin therapy before discharge was necessary. It is necessary for future studies to see all changes involved in antidiabetic treatment in patients with diabetes type 2 and obesity after SARS-CoV2 viral infection and its long-term evolution.
Collapse
Affiliation(s)
- Patricia-Andrada Reștea
- Department of Preclinical Discipline, Doctoral School of Biomedical Science, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Mariana Mureșan
- Department of Preclinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Adrian Voicu
- Department II, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Tunde Jurca
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Annamaria Pallag
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Laura Grațiela Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Ionuț I Jeican
- Department of Morphological Sciences, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Carmen-Bianca Crivii
- Department of Morphological Sciences, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| |
Collapse
|
50
|
Vijakumaran U, Shanmugam J, Heng JW, Azman SS, Yazid MD, Haizum Abdullah NA, Sulaiman N. Effects of Hydroxytyrosol in Endothelial Functioning: A Comprehensive Review. Molecules 2023; 28:molecules28041861. [PMID: 36838850 PMCID: PMC9966213 DOI: 10.3390/molecules28041861] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Pharmacologists have been emphasizing and applying plant and herbal-based treatments in vascular diseases for decades now. Olives, for example, are a traditional symbol of the Mediterranean diet. Hydroxytyrosol is an olive-derived compound known for its antioxidant and cardioprotective effects. Acknowledging the merit of antioxidants in maintaining endothelial function warrants the application of hydroxytyrosol in endothelial dysfunction salvage and recovery. Endothelial dysfunction (ED) is an impairment of endothelial cells that adversely affects vascular homeostasis. Disturbance in endothelial functioning is a known precursor for atherosclerosis and, subsequently, coronary and peripheral artery disease. However, the effects of hydroxytyrosol on endothelial functioning were not extensively studied, limiting its value either as a nutraceutical supplement or in clinical trials. The action of hydroxytyrosol in endothelial functioning at a cellular and molecular level is gathered and summarized in this review. The favorable effects of hydroxytyrosol in the improvement of endothelial functioning from in vitro and in vivo studies were scrutinized. We conclude that hydroxytyrosol is capable to counteract oxidative stress, inflammation, vascular aging, and arterial stiffness; thus, it is beneficial to preserve endothelial function both in vitro and in vivo. Although not specifically for endothelial dysfunction, hydroxytyrosol safety and efficacy had been demonstrated via in vivo and clinical trials for cardiovascular-related studies.
Collapse
|