1
|
Raza ML, Imam MH, Zehra W, Jamil S. Neuro-inflammatory pathways in COVID-19-induced central nervous system injury: Implications for prevention and treatment strategies. Exp Neurol 2024; 382:114984. [PMID: 39368535 DOI: 10.1016/j.expneurol.2024.114984] [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: 06/17/2024] [Revised: 09/15/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
This review explores the neuroinflammatory pathways underlying COVID-19-induced central nervous system (CNS) injury, with a focus on mechanisms of brain damage and strategies for prevention. A comprehensive literature review was conducted to summarize current knowledge on the pathways by which SARS-CoV-2 reaches the brain, the neuroinflammatory responses triggered by viral infection, neurological symptoms and long COVID. Results: We discuss the mechanisms of neuroinflammation in COVID-19, including blood-brain barrier disruption, cytokine storm, microglial activation, and peripheral immune cell infiltration. Additionally, we highlight potential strategies for preventing CNS injury, including pharmacological interventions, immunomodulatory therapies, and lifestyle modifications. Conclusively, Understanding the neuroinflammatory pathways in COVID-19-induced CNS injury is crucial for developing effective prevention and treatment strategies to protect brain health during and after viral infection.
Collapse
Affiliation(s)
- Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | | | | | - Subia Jamil
- Faculty of Pharmacy, Jinnah University for Women, University, Karachi, Pakistan
| |
Collapse
|
2
|
Ullah S, Rahman W, Ullah F, Ullah A, Jehan R, Iqbal MN, Irfan M. A molecular dynamics simulations analysis of repurposing drugs for COVID-19 using bioinformatics methods. J Biomol Struct Dyn 2024; 42:9561-9570. [PMID: 37882340 DOI: 10.1080/07391102.2023.2256864] [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: 05/04/2023] [Accepted: 08/20/2023] [Indexed: 10/27/2023]
Abstract
A number of multidisciplinary methods have piqued the interest of researchers as means to accelerate and lower the cost of medication creation. The goal of this research was to find target proteins and then select a lead drug against SARS-CoV-2. The three-dimensional structure is taken from the RCSB PDB using its specific PDB ID 6lu7. Virtual screening based on pharmacophores is performed using Molecular Operating Environment software. We looked for a potent inhibitor in the FDA-approved database. For docking, AutoDock Vina uses Pyrx. The compound-target protein binding interactions were tested using BIOVIA Discovery Studio. The stability of protein and inhibitor complexes in a physiological setting was investigated using Desmond's Molecular Dynamics Simulation (MD simulation). According to our findings, we repurpose the FDA-approved drugs ZINC000169677008 and ZINC000169289767, which inhibit the activity of the virus's main protease (6lu7). The scientific community will gain from this finding, which might create new medicine. The novel repurposed chemicals were promising inhibitors with increased efficacy and fewer side effects.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | | | - Anees Ullah
- S Khan Lab Mardan, Khyber Pakhtunkhwa, Pakistan
| | | | - Muhammad Nasir Iqbal
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Irfan
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
3
|
Abdelazim MH, Alsenani F, Alnuhait M, Alshammari AS, Altemani AH, Althagafi EA, Waggas DS, Abdelazim AH, Alharbi A. Efficacy of forskolin as a promising therapy for chronic olfactory dysfunction post COVID-19. Eur Arch Otorhinolaryngol 2024; 281:5793-5799. [PMID: 39001919 DOI: 10.1007/s00405-024-08802-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: 03/15/2024] [Accepted: 06/20/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE Olfactory dysfunction is increasingly common among COVID-19 patients, impacting their well-being. Reports have demonstrated decreased levels of cyclic adenosine monophosphate and cyclic guanosine monophosphate among patients with chronic olfactory dysfunction. A prospective randomized clinical trial was developed to demonstrate the efficacy of an oral forskolin regimen treatment, an adenylyl cyclase activator that raises intracellular levels of cyclic adenosine monophosphate, for the treatment of olfactory dysfunction following COVID-19, compared to placebo regimen. METHODS The study enrolled 285 participants with persistent olfactory dysfunction post COVID-19 infection, randomly assigning them to receive either placebo capsules (n = 120) or oral forskolin capsules (n = 165). Follow-up was conducted to track progress, with 18 participants from the placebo group and 12 from the forskolin group lost during this period. Olfactory function was assessed using the "Sniffin' Sticks" test, measuring threshold, discrimination and identification scores before and after treatment. RESULTS Subjects administered forskolin capsules demonstrated a significant enhancement in their composite TDI (threshold, discrimination and identification) score, suggesting a notable amelioration in olfactory functionality. Moreover, the discrimination and identification scores notably improved within the forskolin group. Conversely, no significant alterations were observed in the threshold scores. CONCLUSION This study suggests that forskolin can contribute potentially to improve chronic olfactory dysfunction post COVID-19. TRIAL REGISTRATION DFM-IRB00012367-23-10-001.
Collapse
Affiliation(s)
- Mohamed H Abdelazim
- Department of Otolaryngology, Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Faisal Alsenani
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohammed Alnuhait
- Pharmaceutical Practices Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdullah S Alshammari
- Pharmaceutical Practices Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdullah H Altemani
- Department of Family and Community Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Eyad A Althagafi
- Department of pharmaceutical care, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Dania S Waggas
- Department of Pathological Sciences, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Ahmed H Abdelazim
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt.
| | - Adnan Alharbi
- Pharmaceutical Practices Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
4
|
Yun C, Lee Y, Heo SJ, Kim N, Jung I. The impact of COVID-19 status and vaccine type following the first dose on acute heart disease: A nationwide retrospective cohort study in South Korea. Epidemiol Infect 2024; 152:e134. [PMID: 39444354 PMCID: PMC11502425 DOI: 10.1017/s0950268824001213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 10/25/2024] Open
Abstract
Recent studies have suggested an increased incidence of myocarditis and pericarditis following mRNA vaccination or COVID-19. However, the potential interaction effect between vaccine type and COVID-19 on heart disease risk remains uncertain. Our study aimed to examine the impact of COVID-19 status and vaccine type following the first dose on acute heart disease in the Korean population, using data from the National Health Insurance Service COVID-19 database (October 2018-March 2022). We sought to provide insights for public health policies and clinical decisions pertaining to COVID-19 vaccination strategies. We analysed heart disease risk, including acute cardiac injury, acute myocarditis, acute pericarditis, cardiac arrest, and cardiac arrhythmia, in relation to vaccine type and COVID-19 within 21 days after the first vaccination date, employing Cox proportional hazards models with time-varying covariates. This study included 3,350,855 participants. The results revealed higher heart disease risk in individuals receiving mRNA vaccines than other types (adjusted HR, 1.48; 95% CI, 1.35-1.62). Individuals infected by SARS-CoV-2 also exhibited significantly higher heart disease risk than those uninfected (adjusted HR, 3.56; 95% CI, 1.15-11.04). We found no significant interaction effect between vaccine type and COVID-19 status on the risk of acute heart disease. Notably, however, younger individuals who received mRNA vaccines had a higher heart disease risk compared to older individuals. These results may suggest the need to consider alternative vaccine options for the younger population. Further research is needed to understand underlying mechanisms and guide vaccination strategies effectively.
Collapse
Affiliation(s)
- Choa Yun
- Department of Biostatistics and Computing, Yonsei University, Seoul, South Korea
| | - Yaeji Lee
- Department of Biostatistics and Computing, Yonsei University, Seoul, South Korea
| | - Seok-Jae Heo
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea
| | - Namhui Kim
- Department of Biostatistics and Computing, Yonsei University, Seoul, South Korea
| | - Inkyung Jung
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
5
|
Suhandi C, Wilar G, Narsa AC, Mohammed AFA, El-Rayyes A, Muchtaridi M, Shamsuddin S, Safuan S, Wathoni N. Updating the Pharmacological Effects of α-Mangostin Compound and Unraveling Its Mechanism of Action: A Computational Study Review. Drug Des Devel Ther 2024; 18:4723-4748. [PMID: 39469723 PMCID: PMC11514645 DOI: 10.2147/dddt.s478388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024] Open
Abstract
α-Mangostin, initially identified in 1855, is a xanthone derivative compound predominantly located in the pericarp of the mangosteen fruit (Garcinia mangostana L). This compound is known for its beneficial properties as an antioxidant and anti-inflammatory agent, still holding promise for potential benefits in other related pathologies. In the investigative process, computational studies have proven highly valuable in providing evidence and initial screening before progressing to preclinical and clinical studies. This review aims to present the pharmacological findings and mechanisms of action of α-mangostin based on computational studies. The compilation of this review is founded on the analysis of relevant articles obtained from PubMed, Scopus, and ScienceDirect databases. The study commences with an elucidation of the physicochemical characteristics, drug-likeness, pharmacokinetics, and toxicity profile of α-mangostin, which demonstrates that α-mangostin complies with the Lipinski's Rule of Five, exhibits favorable profiles of absorption, distribution, metabolism, and excretion, and presents low toxicity. Subsequent investigations have revealed that computational studies employing various software tools including ArgusLab, AutoDock, AutoDock Vina, Glide, HEX, and MOE, have been pivotal to comprehend the pharmacology of α-mangostin. Beyond its well established roles as an antioxidant and anti-inflammatory agent, α-mangostin is now recognized for its pharmacological effects in Alzheimer's disease, diabetes, cancer, chronic kidney disease, chronic periodontitis, infectious diseases, and rheumatoid arthritis. Moreover, α-mangostin is projected to have applications in pain management and as a potent mosquito larvicide. All of these findings are based on the attainment of adequate binding affinity to specific target receptors associated with each respective pathological condition. Consequently, it is anticipated that these findings will serve as a foundation for future scientific endeavours, encompassing both in vitro and in vivo studies, as well as clinical investigations, to better understand the pharmacological effects of α-mangostin.
Collapse
Affiliation(s)
- Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Angga Cipta Narsa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Mulawarman University, Samarinda, 71157, Indonesia
| | | | - Ali El-Rayyes
- Department of Chemistry, College of Science, Northern Border University, Arar, Saudi Arabia
| | - Muchtaridi Muchtaridi
- Department of Analytical Pharmacy and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Sabreena Safuan
- Department of Biomedicine, School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| |
Collapse
|
6
|
Serwaa A, Oyawoye F, Owusu IA, Dosoo D, Manu AA, Sobo AK, Fosu K, Olwal CO, Quashie PK, Aikins AR. In vitro analysis suggests that SARS-CoV-2 infection differentially modulates cancer-like phenotypes and cytokine expression in colorectal and prostate cancer cells. Sci Rep 2024; 14:24625. [PMID: 39427065 PMCID: PMC11490510 DOI: 10.1038/s41598-024-75718-1] [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: 05/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) reportedly exacerbates cancer outcomes. However, how COVID-19 influences cancer prognosis and development remains poorly understood. Here, we investigated the effect of Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2), the etiological agent of COVID-19, on cellular cancer phenotypes the expression of cancer-related markers, and various proinflammatory cytokines. We infected prostate (22RV1) and colorectal (DLD-1) cancer cell lines, which express angiotensin-converting enzyme 2 (ACE2), with spike pseudovirus (sPV) and laboratory stocks of live SARS-CoV-2 viruses. After infection, we quantified changes in the cellular cancer phenotypes, the gene expression levels of some cancer markers, including Ki-67, BCL-2, VIM, MMP9, and VEGF, and proinflammatory cytokines. Phenotypic analysis was performed using MTT and wound healing assays, whereas gene expression analysis was carried out using real-time quantitative PCR (RT-qPCR). We show that SARS-CoV-2 infection impacts several key cellular pathways involved in cell growth, apoptosis, and migration, in prostate and colorectal cancer cells. Our results suggest that SARS-CoV-2 infection does influence various cancer cellular phenotypes and expression of molecular cancer markers and proinflammatory cytokines, albeit in a cell-type-specific manner. Our findings hint at the need for further studies and could have implications for evaluating the impact of other viruses on cancer progression.
Collapse
Affiliation(s)
- Alberta Serwaa
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Fatima Oyawoye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Irene Amoakoh Owusu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Daniel Dosoo
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Aaron Adom Manu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Augustine Kojo Sobo
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Fosu
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Charles Ochieng Olwal
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Peter Kojo Quashie
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
- Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - Anastasia Rosebud Aikins
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
- Biochemistry, Cell, and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.
| |
Collapse
|
7
|
Wimalawansa SJ. Unveiling the Interplay-Vitamin D and ACE-2 Molecular Interactions in Mitigating Complications and Deaths from SARS-CoV-2. BIOLOGY 2024; 13:831. [PMID: 39452140 PMCID: PMC11504239 DOI: 10.3390/biology13100831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/26/2024]
Abstract
The interaction of the SARS-CoV-2 spike protein with membrane-bound angiotensin-converting enzyme-2 (ACE-2) receptors in epithelial cells facilitates viral entry into human cells. Despite this, ACE-2 exerts significant protective effects against coronaviruses by neutralizing viruses in circulation and mitigating inflammation. While SARS-CoV-2 reduces ACE-2 expression, vitamin D increases it, counteracting the virus's harmful effects. Vitamin D's beneficial actions are mediated through complex molecular mechanisms involving innate and adaptive immune systems. Meanwhile, vitamin D status [25(OH)D concentration] is inversely correlated with severity, complications, and mortality rates from COVID-19. This study explores mechanisms through which vitamin D inhibits SARS-CoV-2 replication, including the suppression of transcription enzymes, reduced inflammation and oxidative stress, and increased expression of neutralizing antibodies and antimicrobial peptides. Both hypovitaminosis D and SARS-CoV-2 elevate renin levels, the rate-limiting step in the renin-angiotensin-aldosterone system (RAS); it increases ACE-1 but reduces ACE-2 expression. This imbalance leads to elevated levels of the pro-inflammatory, pro-coagulatory, and vasoconstricting peptide angiotensin-II (Ang-II), leading to widespread inflammation. It also causes increased membrane permeability, allowing fluid and viruses to infiltrate soft tissues, lungs, and the vascular system. In contrast, sufficient vitamin D levels suppress renin expression, reducing RAS activity, lowering ACE-1, and increasing ACE-2 levels. ACE-2 cleaves Ang-II to generate Ang(1-7), a vasodilatory, anti-inflammatory, and anti-thrombotic peptide that mitigates oxidative stress and counteracts the harmful effects of SARS-CoV-2. Excess ACE-2 molecules spill into the bloodstream as soluble receptors, neutralizing and facilitating the destruction of the virus. These combined mechanisms reduce viral replication, load, and spread. Hence, vitamin D facilitates rapid recovery and minimizes transmission to others. Overall, vitamin D enhances the immune response and counteracts the pathological effects of SARS-CoV-2. Additionally, data suggests that widely used anti-hypertensive agents-angiotensin receptor blockers and ACE inhibitors-may lessen the adverse impacts of SARS-CoV-2, although they are less potent than vitamin D.
Collapse
|
8
|
Yang H, Wang X. Enhancing Predictive Accuracy of Pediatric COVID-19 Mortality: Integrating Clinical Indicators Beyond HCT-ALB [Letter]. Infect Drug Resist 2024; 17:4375-4376. [PMID: 39411500 PMCID: PMC11476366 DOI: 10.2147/idr.s498364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Affiliation(s)
- Huiye Yang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| | - Xiaotao Wang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People’s Republic of China
| |
Collapse
|
9
|
Ciudad CJ, Valiuska S, Rojas JM, Nogales-Altozano P, Aviñó A, Eritja R, Chillón M, Sevilla N, Noé V. Polypurine Reverse Hoogsteen hairpins as a therapeutic tool for SARS-CoV-2 infection. J Biol Chem 2024:107884. [PMID: 39395809 DOI: 10.1016/j.jbc.2024.107884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/14/2024] Open
Abstract
Although COVID-19 pandemic was declared no longer a global emergency by the World Health Organization in May 2023, SARS-CoV-2 is still infecting people across the world. Many therapeutic oligonucleotides such as ASOs, siRNAs or CRISPR-based systems emerged as promising antiviral strategies for the treatment of SARS-CoV-2. In this work we explored the inhibitory potential on SARS-CoV-2 replication of Polypurine Reverse Hoogsteen Hairpins (PPRHs), CC1-PPRH and CC3-PPRH, targeting specific polypyrimidine sequences within the replicase and Spike regions, respectively, and previously validated for COVID-19 diagnosis. Both PPRHs bound to their target sequences in the viral genome with high affinity in the order of nM. In vitro, both PPRHs reduced viral replication by more than 92% when transfected into VERO-E6 cells 24 hours prior infection with SARS-CoV-2. In vivo intranasal administration of CC1-PPRH in K18-hACE2 mice expressing the human ACE receptor protected all the animals from SARS-CoV-2 infection. The properties of PPRHs position them as promising candidates for the development of novel therapeutics against SARS-CoV-2 and other viral infections.
Collapse
Affiliation(s)
- Carlos J Ciudad
- Department of Biochemistry & Physiology, School Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain; Institut de Nanociencia i Nanotecnologia (IN2UB), Universitat de Barcelona Barcelona, Spain.
| | - Simonas Valiuska
- Department of Biochemistry & Physiology, School Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain; Institut de Nanociencia i Nanotecnologia (IN2UB), Universitat de Barcelona Barcelona, Spain
| | - José Manuel Rojas
- Centro de Investigación en Sanidad Animal-CISA, INIA, CSIC, Madrid, Spain
| | | | - Anna Aviñó
- Institute for Advanced Chemistry of Catalonia, CSIC, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
| | - Ramón Eritja
- Institute for Advanced Chemistry of Catalonia, CSIC, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Chillón
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal-CISA, INIA, CSIC, Madrid, Spain
| | - Verónique Noé
- Department of Biochemistry & Physiology, School Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain; Institut de Nanociencia i Nanotecnologia (IN2UB), Universitat de Barcelona Barcelona, Spain
| |
Collapse
|
10
|
Liu Y, Lou X. The Bidirectional Association Between Metabolic Syndrome and Long-COVID-19. Diabetes Metab Syndr Obes 2024; 17:3697-3710. [PMID: 39398386 PMCID: PMC11471063 DOI: 10.2147/dmso.s484733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024] Open
Abstract
Background The rapid global spread of a new coronavirus disease known as COVID-19 has led to a significant increase in mortality rates, resulting in an unprecedented worldwide pandemic. Methods The impact of COVID-19, particularly its long-term effects, has also had a profound effect on the health and well-being of individuals.Metabolic syndrome increases the risk of heart and brain diseases, presenting a significant danger to human well-being. Purpose The prognosis of long COVID and the progression of metabolic syndrome interact with each other, but there is currently a lack of systematic reports.In this paper, the pathogenesis, related treatment and prognosis of long COVID and metabolic syndrome are systematically reviewed.
Collapse
Affiliation(s)
- Yanfen Liu
- Department of Endocrinology at Zhejiang University School of Medicine, Jinhua Hospital, Jinhua, People’s Republic of China
| | - Xueyong Lou
- Department of Endocrinology at Zhejiang University School of Medicine, Jinhua Hospital, Jinhua, People’s Republic of China
| |
Collapse
|
11
|
Fiorucci S, Urbani G, Biagioli M, Sepe V, Distrutti E, Zampella A. Bile acids and bile acid activated receptors in the treatment of Covid-19. Biochem Pharmacol 2024; 228:115983. [PMID: 38081371 DOI: 10.1016/j.bcp.2023.115983] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 09/20/2024]
Abstract
Since its first outbreak in 2020, the pandemic caused by the Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) has caused the death of almost 7 million people worldwide. Vaccines have been fundamental in disease prevention and to reduce disease severity especially in patients with comorbidities. Nevertheless, treatment of COVID-19 has been proven difficult and several approaches have failed to prevent disease onset or disease progression, particularly in patients with comorbidities. Interrogation of drug data bases has been widely used since the beginning of pandemic to repurpose existing drugs/natural substances for the prevention/treatment of COVID-19. Steroids, including bile acids such as ursodeoxycholic acid (UDCA) and chenodeoxycholic acid (CDCA) have shown to be promising for their potential in modulating SARS-CoV-2/host interaction. Bile acids have proven to be effective in preventing binding of spike protein with the Angiotensin Converting Enzyme II (ACE2), thus preventing virus uptake by the host cells and inhibiting its replication, as well as in indirectly modulating immune response. Additionally, the two main bile acid activated receptors, GPBAR1 and FXR, have proven effective in modulating the expression of ACE2, suggesting an indirect role for these receptors in regulating SARS-CoV-2 infectiveness and immune response. In this review we have examined how the potential of bile acids and their receptors as anti-COVID-19 therapies and how these biochemical mechanisms translate into clinical efficacy.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Ginevra Urbani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Valentina Sepe
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Angela Zampella
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
12
|
Chang K, Ahorsu DK, Tsai H, Strong C, Ko N, Chen J, Yen C, Üztemur S, Griffiths MD, Lin C. Parallel Mediating Effects of Sleep Quality, Psychological Distress, and Self-Stigma in the Associations Between Long COVID Symptoms and Quality of Life Among Taiwanese Individuals With Mental Health Illness. Brain Behav 2024; 14:e70094. [PMID: 39402811 PMCID: PMC11473576 DOI: 10.1002/brb3.70094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/27/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Long COVID symptoms (i.e., experiencing symptoms of COVID-19 for 3 months post-COVID-19) affect individuals' health and their quality of life (QoL). However, the pathways through which it does so are not fully known. AIM The present study examined the mediating roles of sleep quality, psychological distress, and self-stigma in the associations between long COVID symptoms and QoL among individuals with mental illness. METHOD Individuals with mental illness (n = 333) were recruited from a psychiatric center in southern Taiwan to participate in the study. Data were collected regarding sleep quality, psychological distress, self-stigma, and QoL. Independent t-tests, Pearson correlations, and regression with Hayes' Process Macro were used to compare groups, examine relationships, and parallel mediation models, respectively. RESULTS Participants with long COVID symptoms had significantly worse sleep quality, psychological distress, physical QoL, and psychological QoL compared to those without symptoms. There were significant relationships between sleep quality, psychological distress, self-stigma, and QoL. Sleep quality significantly mediated the associations between long COVID symptoms and physical and social QoL. Psychological distress significantly mediated the associations between long COVID symptoms and all domains of QoL, but not self-stigma. CONCLUSION There are alternative pathways (e.g., sleep quality and psychological distress) through which long COVID symptoms may affect the QoL of individuals with mental illness. The findings suggest that individuals with long COVID symptoms have a higher chance of having poor QoL. Therefore, there may be the need for counseling and possible therapy for those who contract COVID-19, especially among individuals who already have mental illness.
Collapse
Affiliation(s)
- Kun‐Chia Chang
- Department of General Psychiatry, Jianan Psychiatric CenterMinistry of Health and WelfareTainanTaiwan
- Department of Psychiatry, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Daniel Kwasi Ahorsu
- Department of Special Education and CounsellingThe Education University of Hong KongTai PoNew TerritoriesHong Kong
| | - Hsin‐Chi Tsai
- Department of Psychiatry, School of MedicineTzu Chi UniversityHualienTaiwan
- Department of PsychiatryTzu‐Chi General HospitalHualienTaiwan
| | - Carol Strong
- Department of Public Health, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Nai‐Ying Ko
- Department of Nursing, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Jung‐Sheng Chen
- Department of Medical ResearchE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
| | - Cheng‐Fang Yen
- Department of PsychiatryKaohsiung Medical University HospitalKaohsiungTaiwan
- Department of Psychiatry, School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- College of Professional StudiesNational Pingtung University of Science and TechnologyPingtungTaiwan
| | - Servet Üztemur
- Department of Turkish and Social Sciences Education, Faculty of EducationAnadolu UniversityEskişehirTürkiye
| | | | - Chung‐Ying Lin
- Department of Public Health, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Institute of Allied Health Sciences, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Biostatistics Consulting Center, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Department of Occupational Therapy, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
13
|
He C, Li R, Zhang J, Chai W. Sivelestat protects against acute lung injury by up-regulating angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas receptors. J Thorac Dis 2024; 16:6182-6195. [PMID: 39444885 PMCID: PMC11494560 DOI: 10.21037/jtd-24-1281] [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: 08/07/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Background Acute lung injury (ALI) and its most severe manifestation of acute respiratory distress syndrome (ARDS) is a disease with a clinical mortality rate of up to 40% and is one of the most dangerous and common complications of severe coronavirus disease 2019 (COVID-19). Sivelestat (SIV) is the only licensed therapeutic medicine in the world for ALI/ARDS treatment. The angiotensin-converting enzyme 2 (ACE2)/angiotensin (Ang)-(1-7)/Mas receptor axis is critical in the prevention of ALI/ARDS. This study aims to investigate whether SIV alleviates lipopolysaccharides (LPS)-induced ALI by inhibiting the down-regulation of ACE2/Ang-(1-7)/Mas receptor axis expression. Methods In vivo, 90 male Sprague-Dawley rats were randomized into 5 groups. Then, we pretreated different groups of rats with dexamethasone (DEX) or SIV. Rats were sacrificed at three different time points (3, 6, and 12 hours) following LPS instillation. In vitro, RAW264.7 cells were divided into 11 groups. Different groups of cells were pretreated with DEX or SIV. And then added with LPS for 3, 6, and 12 hours. Next, we introduced A779, a potent Ang-(1-7) receptor antagonist, and DX600 as the ACE2 antagonist in different groups. Then the protein and messenger RNA (mRNA) expression levels of ACE2 in rat lung tissue and the expression levels of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and Ang-(1-7) in the rat serum and the cell culture supernatant were measured. And the data were statistically analyzed. Results In vivo, the rats pretreated with SIV or DEX had significantly lower lung wet/dry (W/D) ratios and lung pathological alterations than those exposed to LPS only. Both in vivo and in vitro, we observed that SIV or DEX significantly attenuated the LPS-induced up-regulation of IL-6 and TNF-α levels, and the down-regulation of ACE2 and Ang-(1-7) levels. In vitro, the pretreatment of the RAW264.7 cells with DX600 and A779 significantly reduced and even abolished the protective effects of SIV. Conclusions Therefore, it was concluded that SIV protected against LPS-induced ALI and decreased inflammatory cytokine release by up-regulating the ACE2/Ang-(1-7)/Mas receptor axis. Our results enrich the theoretical foundation for the clinical application of SIV and provide fresh ideas for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Changqing He
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ruoxin Li
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jia Zhang
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Wenshu Chai
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
14
|
Seethamraju H, Yang OO, Loftus R, Ogbuagu O, Sammartino D, Mansour A, Sacha JB, Ojha S, Hansen SG, Arman AC, Lalezari JP. A Randomized Placebo-Controlled Trial of Leronlimab in Mild-To-Moderate COVID-19. Clin Ther 2024:S0149-2918(24)00260-1. [PMID: 39353749 DOI: 10.1016/j.clinthera.2024.08.019] [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: 04/24/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE Early in the course of the SARS-CoV-2 pandemic it was hypothesised that host genetics played a role in the pathophysiology of COVID-19 including a suggestion that the CCR5-Δ32 mutation may be protective in SARS-CoV-2 infection. Leronlimab is an investigational CCR5-specific humanized IgG4 monoclonal antibody currently in development for HIV-1 infection. We aimed to explore the impact of leronlimab on the severity of disease symptoms among participants with mild-to-moderate COVID-19. METHODS The TEMPEST trial was a randomized, double-blind, placebo-controlled study in participants with mild-to-moderate COVID-19. Participants were randomly assigned in a 2:1 ratio to receive subcutaneous leronlimab (700 mg) or placebo on days 0 and 7. The primary efficacy endpoint was assessed by change in total symptom score based on fever, myalgia, dyspnea, and cough, at end of treatment (day 14). FINDINGS Overall, 84 participants were randomized and treated with leronlimab (n = 56) or placebo (n = 28). No difference was observed in change in total symptom score (P = 0.8184) or other pre-specified secondary endpoints between treatments. However, in a post hoc analysis, 50.0% of participants treated with leronlimab demonstrated improvements from baseline in National Early Warning Score 2 (NEWS2) at day 14, compared with 20·8% of participants in the placebo group (post hoc; p = 0.0223). Among participants in this trial with mild-to-moderate COVID-19 adverse events rates were numerically but not statistically significantly lower in leronlimab participants (33.9%) compared with placebo participants (50.0%). IMPLICATIONS At the time the TEMPEST trial was designed although CCR5 was known to be implicated in COVID-19 disease severity the exact pathophysiology of SARS-CoV-2 infection was poorly understood. Today it is well accepted that SARS-CoV-2 infection in asymptomatic-to-mild cases is primarily characterized by viral replication, with a heightened immune response, accompanied by diminished viral replication in moderate-to-severe disease and a peak in inflammatory responses with excessive production of pro-inflammatory cytokines in critical disease. It is therefore perhaps not surprising that no differences between treatments were observed in the primary endpoint or in pre-specified secondary endpoints among participants with mild-to-moderate COVID-19. However, the results of the exploratory post hoc analysis showing that participants in the leronlimab group had greater improvement in NEWS2 assessment compared to placebo provided a suggestion that leronlimab may be associated with a lower likelihood of people with mild-to-moderate COVID-19 progressing to more severe disease and needs to be confirmed in other appropriately designed clinical trials. CLINICALTRIALS gov number, NCT04343651 https://classic. CLINICALTRIALS gov/ct2/show/NCT04343651.
Collapse
Affiliation(s)
| | - Otto O Yang
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | | | | | | | - Jonah B Sacha
- Oregon Health & Science University, Portland, Oregon
| | - Sohita Ojha
- Oregon Health & Science University, Portland, Oregon
| | | | | | | |
Collapse
|
15
|
Kruger A, Joffe D, Lloyd-Jones G, Khan MA, Šalamon Š, Laubscher GJ, Putrino D, Kell DB, Pretorius E. Vascular Pathogenesis in Acute and Long COVID: Current Insights and Therapeutic Outlook. Semin Thromb Hemost 2024. [PMID: 39348850 DOI: 10.1055/s-0044-1790603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Long coronavirus disease 2019 (COVID-19)-a postacute consequence of severe acute respiratory syndrome coronavirus 2 infection-manifests with a broad spectrum of relapsing and remitting or persistent symptoms as well as varied levels of organ damage, which may be asymptomatic or present as acute events such as heart attacks or strokes and recurrent infections, hinting at complex underlying pathogenic mechanisms. Central to these symptoms is vascular dysfunction rooted in thrombotic endothelialitis. We review the scientific evidence that widespread endothelial dysfunction (ED) leads to chronic symptomatology. We briefly examine the molecular pathways contributing to endothelial pathology and provide a detailed analysis of how these cellular processes underpin the clinical picture. Noninvasive diagnostic techniques, such as flow-mediated dilation and peripheral arterial tonometry, are evaluated for their utility in identifying ED. We then explore mechanistic, cellular-targeted therapeutic interventions for their potential in treating ED. Overall, we emphasize the critical role of cellular health in managing Long COVID and highlight the need for early intervention to prevent long-term vascular and cellular dysfunction.
Collapse
Affiliation(s)
- Arneaux Kruger
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - David Joffe
- Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, Australia
- World Health Network, Cambridge, Massachusetts
| | - Graham Lloyd-Jones
- Department of Radiology, Salisbury District Hospital, Salisbury NHS Foundation Trust, United Kingdom
| | - Muhammed Asad Khan
- World Health Network, Cambridge, Massachusetts
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | | | | | - David Putrino
- Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, Australia
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- World Health Network, Cambridge, Massachusetts
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
16
|
Gullo G, Lopez A, Loreto C, Cucinella G, La Verde M, Andrisani A, Burgio S, Carotenuto R, Ganduscio S, Baglio G, Billone V, Perino A, De Franciscis P, Marinelli S. COVID-19 and Female Fertility: An Observational Prospective Multicenter Cohort Study: Upholding Reproductive Rights in Emergency Circumstances. Diagnostics (Basel) 2024; 14:2118. [PMID: 39410522 PMCID: PMC11475084 DOI: 10.3390/diagnostics14192118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
OBJECTIVES Currently available research data points to COVID-19-related multi-organ system damage. This study aims to evaluate the impact of SARS-CoV-2 on the reproductive health, that is, plasma levels of FSH, LH, estradiol, AMH, and antral follicular count, of women undergoing level II ART techniques. METHODS This is a multicenter, prospective, and observational study by the reproductive medicine centers of Palermo's Ospedali Riuniti Villa Sofia-Cervello Hospital and Vanvitelli University. From September 2022 to March 2024, 203 patients aged 24-43 were enrolled, all with diagnosed infertility and a history of SARS-CoV-2 infection. Symptomatic women, patients testing positive for HIV or other liver viruses, and patients with a history of ovarian cancer or who had taken gonadotoxic drugs were excluded. Plasma measurements of FSH, LH, estradiol, AMH, and antral follicular count were performed before and after infection. RESULTS The analysis accounting for the concentration of anti-Müllerian hormone (AMH) before and after COVID-19 infection shows an average concentration decrease from 1.33 ng/mL before SARS-CoV-2 infection to 0.97 ng/mL after infection. Average decrease after infection was -27.4%; average reduction of 1 follicle (95% CI: from -0.74 to -1.33) was reported following SARS-CoV-2 infection. Levels of E2 before and after SARS-CoV-2 infection did not vary significantly. Average FSH and LH levels before and after SARS-CoV-2 infection pointed to an increase. CONCLUSIONS SARS-CoV-2 infection damages female reproductive health, causing significant reductions in AMH (-27.4%) and AFC (-1 antral follicle) values and an increase in FSH (+13.6%) and LH (+13.4%) values. No effect on E2 levels was reported. The pandemic has also affected the ability of infertile patients to access ART procedures, and that calls for a novel, updated blueprint designed to enhance our preparedness in the event that similar circumstances should occur again.
Collapse
Affiliation(s)
- Giuseppe Gullo
- Department of Obstetrics and Gynecology, IVF Unit, AOOR Villa Sofia Cervello, University of Palermo, 90146 Palermo, Italy; (A.L.); (G.C.); (S.G.); (V.B.); (A.P.)
| | - Alessandra Lopez
- Department of Obstetrics and Gynecology, IVF Unit, AOOR Villa Sofia Cervello, University of Palermo, 90146 Palermo, Italy; (A.L.); (G.C.); (S.G.); (V.B.); (A.P.)
| | - Carla Loreto
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80121 Naples, Italy; (C.L.); (M.L.V.); (R.C.); (P.D.F.)
| | - Gaspare Cucinella
- Department of Obstetrics and Gynecology, IVF Unit, AOOR Villa Sofia Cervello, University of Palermo, 90146 Palermo, Italy; (A.L.); (G.C.); (S.G.); (V.B.); (A.P.)
| | - Marco La Verde
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80121 Naples, Italy; (C.L.); (M.L.V.); (R.C.); (P.D.F.)
| | - Alessandra Andrisani
- Unit of Gynecology and Obstetrics, Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy;
| | - Sofia Burgio
- Department of Obstetrics and Gynecology, IVF Unit, AOOR Villa Sofia Cervello, University of Palermo, 90146 Palermo, Italy; (A.L.); (G.C.); (S.G.); (V.B.); (A.P.)
| | - Raffaela Carotenuto
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80121 Naples, Italy; (C.L.); (M.L.V.); (R.C.); (P.D.F.)
| | - Silvia Ganduscio
- Department of Obstetrics and Gynecology, IVF Unit, AOOR Villa Sofia Cervello, University of Palermo, 90146 Palermo, Italy; (A.L.); (G.C.); (S.G.); (V.B.); (A.P.)
| | - Giovanni Baglio
- Italian National Agency for Regional Healthcare Services, 00187 Roma, Italy;
| | - Valentina Billone
- Department of Obstetrics and Gynecology, IVF Unit, AOOR Villa Sofia Cervello, University of Palermo, 90146 Palermo, Italy; (A.L.); (G.C.); (S.G.); (V.B.); (A.P.)
| | - Antonio Perino
- Department of Obstetrics and Gynecology, IVF Unit, AOOR Villa Sofia Cervello, University of Palermo, 90146 Palermo, Italy; (A.L.); (G.C.); (S.G.); (V.B.); (A.P.)
| | - Pasquale De Franciscis
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80121 Naples, Italy; (C.L.); (M.L.V.); (R.C.); (P.D.F.)
| | - Susanna Marinelli
- School of Law, Polytechnic University of Marche, 60121 Ancona, Italy;
| |
Collapse
|
17
|
Lin X, Zhuang Y, Gao F. ACE2 Alleviates Endoplasmic Reticulum Stress and Protects against Pyroptosis by Regulating Ang1-7/Mas in Ventilator-Induced Lung Injury. FRONT BIOSCI-LANDMRK 2024; 29:334. [PMID: 39344337 DOI: 10.31083/j.fbl2909334] [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: 11/06/2023] [Revised: 12/19/2023] [Accepted: 02/04/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Ventilator-induced lung injury (VILI) is a consequence of inflammation and increased alveolar-capillary membrane permeability due to alveolar hyperdistention or elevated intrapulmonary pressure, but the precise mechanisms remain unclear. The aim of the study was to analyze the mechanism by which angiotensin converting enzyme 2 (ACE2) alleviates endoplasmic reticulum stress (ERS) and protects alveolar cells from pyroptosis in VILI by regulating angiotensin (Ang)1-7/Mas. METHODS VILI was induced in mice by mechanical ventilation by regulating the tidal volume. The alveolar cell line, A549, mimics VILI in vitro by cyclic stretch (CS). Ang (1-7) (100 nmol/L) was added to the medium. ERS was induced in cells by stimulating with tunicamycin (TM, 2 μg/mL). ERS was inhibited by tracheal instillation of 4-phenylbutyric acid (4-PBA) (1 mg/kg). ACE2's enzymatic function was activated or inhibited by subcutaneous injection of resorcinolnaphthalein (RES, 20 μg/kg) or MLN-4760 (20 μg/kg). pGLV-EF1a-GFP-ACE2 was instilled into the trachea to increase the protein expression of ACE2. The Ang (1-7) receptor, Mas, was antagonized by injecting A779 subcutaneously (80 μg/kg). RESULTS ACE2 protein levels decreased after modeling. Ang (1-7) level was decreased and Ang II was accumulated. ERS was significantly induced in VILI mice, and pyroptosis was observed in cells. When ERS was inhibited, pyroptosis under the VILI condition was significantly inhibited. Ang (1-7) alleviated ERS and pyroptosis under CS. When ERS was continuously activated, the function of Ang (1-7) in inhibiting pyroptosis was blocked. Resorcinolnaphthalein (RES) effectively promoted Ang II conversion, alleviated the Ang (1-7) level in VILI, ameliorated lung injury, and inhibited ERS and cell pyroptosis. Inhibiting ACE2's function in VILI hindered the production of Ang (1-7), promoted the accumulation of Ang II, and exacerbated ERS and pyroptosis, along with lung injury. The Mas antagonist significantly blocked the inhibitory effects of ACE2 on ERS and pyroptosis in VILI. CONCLUSIONS Reduced ACE2 expression in VILI is involved in ERS and pyroptosis-related injury. ACE2 can alleviate ERS in alveolar cells by catalyzing the production of Ang (1-7), thus inhibiting pyroptosis in VILI.
Collapse
Affiliation(s)
- Xingsheng Lin
- Department of Intensive Care Unit, Fuzhou University Affiliated Provincial Hospital, 350001 Fuzhou, Fujian, China
| | - Yingfeng Zhuang
- Department of Intensive Care Unit, Fuzhou University Affiliated Provincial Hospital, 350001 Fuzhou, Fujian, China
| | - Fengying Gao
- Department of Pulmonary Disease, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 200071 Shanghai, China
| |
Collapse
|
18
|
Gawande V, Kushwaha R, Mandal AA, Banerjee S. Targeting SARS-CoV-2 Proteins: In Silico Investigation with Polypyridyl-Based Zn(II)-Curcumin Complexes. Chembiochem 2024:e202400612. [PMID: 39264259 DOI: 10.1002/cbic.202400612] [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: 07/21/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/13/2024]
Abstract
Herein, we have selected eight Zn(II)-based complexes viz., [Zn(bpy)(acac)Cl] (1), [Zn(phen)(acac)Cl] (2), [Zn(dppz)(acac)Cl] (3), [Zn(dppn)(acac)Cl] (4), [Zn(bpy)(cur)Cl] (5), [Zn(phen)(cur)Cl] (6), [Zn(dppz)(cur)Cl] (7), [Zn(dppn)(cur)Cl] (8), where bpy=2,2'-bipyridine, phen=1,10-phenanthroline, dppz=benzo[i]dipyrido[3,2-a:2',3'-c]phenazine, dppn=naphtho[2,3-i]dipyrido[3,2-a:2',3'-c]phenazine, acac=acetylacetonate, cur=curcumin and performed in silico molecular docking studies with the viral proteins, i. e., spike protein (S), Angiotensin-converting enzyme II Receptor protein (ACE2), nucleocapsid protein (N), main protease protein (Mpro), and RNA-dependent RNA polymerase protein (RdRp) of SARS-CoV-2. The binding energy calculations, visualization of the docking orientation, and analysis of the interactions revealed that these complexes could be potential inhibitors of the viral proteins. Among complexes 1-8, complex 6 showed the strongest binding affinity with S and ACE2 proteins. 4 exerted better binding affinity in the case of the N protein, whereas 8 presented the highest binding affinities with Mpro and RdRp among all the complexes. Overall, the study indicated that Zn(II) complexes have the potential as alternative and viable therapeutic solutions for COVID-19.
Collapse
Affiliation(s)
- Vedant Gawande
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
19
|
Zlosa M, Grubišić B, Švitek L, Sabadi D, Canecki-Varžić S, Mihaljević I, Bilić-Ćurčić I, Kizivat T. Implications of Dysnatremia and Endocrine Disturbances in COVID-19 Patients. Int J Mol Sci 2024; 25:9856. [PMID: 39337343 PMCID: PMC11432667 DOI: 10.3390/ijms25189856] [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: 06/30/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Sodium imbalance is a common electrolyte disturbance in COVID-19, often linked to disruptions in hormonal regulation. This review explores the relationship between sodium dysregulation and endocrine disturbances, particularly focusing on primary and secondary hypothyroidism, hypocortisolism, and the renin-angiotensin-aldosterone system (RAAS). Hypocortisolism in COVID-19, due to adrenal insufficiency or secondary to pituitary dysfunction, can lead to hyponatremia through inadequate cortisol levels, which impair renal free water excretion and enhance antidiuretic hormone (ADH) secretion. Similarly, hypothyroidism is associated with decreased renal blood flow and the glomerular filtration rate (GFR), which also increases ADH activity, leading to water retention and dilutional hyponatremia. Furthermore, COVID-19 can disrupt RAAS (primarily through its interaction with the angiotensin-converting enzyme 2 (ACE2) receptor), diminishing aldosterone secretion and further contributing to sodium loss and hyponatremia. These hormonal disruptions suggest that sodium imbalance in COVID-19 is multifactorial and warrants further investigation into the complex interplay between COVID-19, endocrine function, and sodium homeostasis. Future research should focus on understanding these mechanisms to develop management algorithms that address both sodium imbalance and underlying hormonal disturbances in order to improve prognosis and outcomes in COVID-19 patients.
Collapse
Affiliation(s)
- Mihaela Zlosa
- Clinic for Infectious Diseases, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia; (M.Z.); (B.G.); (D.S.)
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Barbara Grubišić
- Clinic for Infectious Diseases, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia; (M.Z.); (B.G.); (D.S.)
- Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Luka Švitek
- Clinic for Infectious Diseases, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia; (M.Z.); (B.G.); (D.S.)
- Department of Infectology and Dermatovenerology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Dario Sabadi
- Clinic for Infectious Diseases, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia; (M.Z.); (B.G.); (D.S.)
- Department of Infectology and Dermatovenerology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
- Faculty of Dental Medicine and Health Osijek, J. J. Strossmayer University of Osijek, 21 Crkvena Street, HR-31000 Osijek, Croatia
| | - Silvija Canecki-Varžić
- Department of Endocrinology, Internal Medicine Clinic, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia;
- Department of Pathophysiology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Ivica Mihaljević
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia; (I.M.); (T.K.)
- Department for Nuclear Medicine and Oncology, Faculty of Medicine, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
- Academy of Medical Sciences of Croatia, 15 Kaptol Street, HR-10000 Zagreb, Croatia
| | - Ines Bilić-Ćurčić
- Department of Endocrinology, Internal Medicine Clinic, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia;
- Department of Pharmacology, Faculty of Medicine Osijek, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| | - Tomislav Kizivat
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Centre Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia; (I.M.); (T.K.)
- Department for Nuclear Medicine and Oncology, Faculty of Medicine, J. J. Strossmayer University of Osijek, 4 Josip Huttler Street, HR-31000 Osijek, Croatia
| |
Collapse
|
20
|
Tuttolomondo M, Pham STD, Terp MG, Cendán Castillo V, Kalisi N, Vogel S, Langkjær N, Hansen UM, Thisgaard H, Schrøder HD, Palarasah Y, Ditzel HJ. A novel multitargeted self-assembling peptide-siRNA complex for simultaneous inhibition of SARS-CoV-2-host cell interaction and replication. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102227. [PMID: 38939051 PMCID: PMC11203390 DOI: 10.1016/j.omtn.2024.102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
Effective therapeutics are necessary for managing severe COVID-19 disease despite the availability of vaccines. Small interfering RNA (siRNA) can silence viral genes and restrict SARS-CoV-2 replication. Cell-penetrating peptides is a robust method for siRNA delivery, enhancing siRNA stability and targeting specific receptors. We developed a peptide HE25 that blocks SARS-CoV-2 replication by various mechanisms, including the binding of multiple receptors involved in the virus's internalization, such as ACE2, integrins and NRP1. HE25 not only acts as a vehicle to deliver the SARS-CoV-2 RNA-dependent RNA polymerase siRNA into cells but also facilitates their internalization through endocytosis. Once inside endosomes, the siRNA is released into the cytoplasm through the Histidine-proton sponge effect and the selective cleavage of HE25 by cathepsin B. These mechanisms effectively inhibited the replication of the ancestral SARS-CoV-2 and the Omicron variant BA.5 in vitro. When HE25 was administered in vivo, either by intravenous injection or inhalation, it accumulated in lungs, veins and arteries, endothelium, or bronchial structure depending on the route. Furthermore, the siRNA/HE25 complex caused gene silencing in lung cells in vitro. The SARS-CoV-2 siRNA/HE25 complex is a promising therapeutic for COVID-19, and a similar strategy can be employed to combat future emerging viral diseases.
Collapse
Affiliation(s)
- Martina Tuttolomondo
- Department of Molecular Medicine, Unit of Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Stephanie Thuy Duong Pham
- Department of Molecular Medicine, Unit of Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Mikkel Green Terp
- Department of Molecular Medicine, Unit of Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Virginia Cendán Castillo
- Department of Molecular Medicine, Unit of Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Nazmie Kalisi
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5000 Odense, Denmark
| | - Stefan Vogel
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5000 Odense, Denmark
| | - Niels Langkjær
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Ulla Melchior Hansen
- Department of Molecular Medicine, Imaging Core Facility, DaMBIC, University of Southern Denmark, 5000 Odense, Denmark
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Henrik Daa Schrøder
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark
| | - Yaseelan Palarasah
- Department of Molecular Medicine, Unit of Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Henrik Jørn Ditzel
- Department of Molecular Medicine, Unit of Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Department of Oncology, Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
21
|
Piza PMDT, de Freitas VM, Aguiar-Brito I, Calsolari-Oliveira BM, Rangel ÉB. Impact of Hyponatremia on COVID-19-Related Outcomes: A Retrospective Analysis. Biomedicines 2024; 12:1997. [PMID: 39335510 PMCID: PMC11444129 DOI: 10.3390/biomedicines12091997] [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: 07/26/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Sodium disturbances are observed in one-third of patients with COVID-19 and result from multifaceted mechanisms. Notably, hyponatremia is associated with disease progression and mortality. AIM We aimed to analyze the impact of hyponatremia on COVID-19 outcomes and its correlation with clinical and laboratory parameters during the first wave. METHODS We evaluated the sodium levels of 558 patients with COVID-19 between 21 March 2020, and 31 July 2020, at a single center. We performed linear regression analyses to explore the correlation of sodium levels with COVID-19-related outcomes, demographic data, signs and symptoms, and laboratory parameters. Next, we conducted Pearson correlation analyses. A p-value < 0.05 was considered significant. RESULTS Hyponatremia was found in 35.3% of hospitalized patients with COVID-19. This was associated with the need for intensive care transfer (B = -1.210, p = 0.009) and invasive mechanical ventilation (B = -1.063, p = 0.032). Hyponatremia was frequently found in oncologic patients (p = 0.002) and solid organ transplant recipients (p < 0.001). Sodium was positively associated with diastolic blood pressure (p = 0.041) and productive cough (p = 0.022) and negatively associated with dry cough (p = 0.032), anorexia (p = 0.004), and nausea/vomiting (p = 0.007). Regarding the correlation of sodium levels with other laboratory parameters, we observed a positive correlation with hematocrit (p = 0.011), lymphocytes (p = 0.010), pCO2 (p < 0.0001), bicarbonate (p = 0.0001), and base excess (p = 0.008) and a negative correlation with the neutrophil-to-lymphocyte ratio (p = 0.009), the platelet-to-lymphocyte ratio (p = 0.033), and arterial blood glucose (p = 0.016). CONCLUSIONS Hyponatremia is a risk factor for adverse outcomes in COVID-19 patients. It is associated with demographic data and clinical and laboratory parameters. Therefore, hyponatremia is an important tool for risk stratification in COVID-19 patients.
Collapse
Affiliation(s)
- Pedro Maciel de Toledo Piza
- Paulista School of Medicine, Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (P.M.d.T.P.); (V.M.d.F.); (I.A.-B.); (B.M.C.-O.)
| | - Victor Muniz de Freitas
- Paulista School of Medicine, Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (P.M.d.T.P.); (V.M.d.F.); (I.A.-B.); (B.M.C.-O.)
| | - Isabella Aguiar-Brito
- Paulista School of Medicine, Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (P.M.d.T.P.); (V.M.d.F.); (I.A.-B.); (B.M.C.-O.)
| | - Barbara Monique Calsolari-Oliveira
- Paulista School of Medicine, Federal University of São Paulo, São Paulo 04023-062, SP, Brazil; (P.M.d.T.P.); (V.M.d.F.); (I.A.-B.); (B.M.C.-O.)
| | - Érika Bevilaqua Rangel
- Department of Medicine, Nephrology Division, Federal University of São Paulo, São Paulo 04038-031, SP, Brazil
- Instituto Israelita de Ensino e Pesquisa Albert Einstein, Hospital Israelita Albert Einstein, São Paulo 05652-900, SP, Brazil
| |
Collapse
|
22
|
Oliveira EH, Monteleone-Cassiano AC, Tavares L, Santos JC, Lima TM, Gomes GF, Tanaka PP, Monteiro CJ, Munuera M, Batah SS, Fabro AT, Faça VM, Masson AP, Donadi EA, Dametto M, Bonacin R, Martins RB, Neto EA, daSilva LLP, Cunha TM, Passos GA. A mimetic peptide of ACE2 protects against SARS-CoV-2 infection and decreases pulmonary inflammation related to COVID-19. Antiviral Res 2024; 229:105968. [PMID: 39004311 DOI: 10.1016/j.antiviral.2024.105968] [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: 04/15/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Since human angiotensin-converting enzyme 2 (ACE2) serves as a primary receptor for SARS-CoV-2, characterizing ACE2 regions that allow SARS-CoV-2 to enter human cells is essential for designing peptide-based antiviral blockers and elucidating the pathogenesis of the virus. We identified and synthesized a 25-mer mimetic peptide (encompassing positions 22-46 of the ACE2 alpha-helix α1) implicated in the S1 receptor-binding domain (RBD)-ACE2 interface. The mimetic (wild-type, WT) ACE2 peptide significantly inhibited SARS-CoV-2 infection of human pulmonary Calu-3 cells in vitro. In silico protein modeling predicted that residues F28, K31, F32, F40, and Y41 of the ACE2 alpha-helix α1 are critical for the original, Delta, and Omicron strains of SARS-CoV-2 to establish the Spike RBD-ACE2 interface. Substituting these residues with alanine (A) or aspartic acid (D) abrogated the antiviral protective effect of the peptides, indicating that these positions are critical for viral entry into pulmonary cells. WT ACE2 peptide, but not the A or D mutated peptides, exhibited significant interaction with the SARS-CoV-2 S1 RBD, as shown through molecular dynamics simulations. Through identifying the critical amino acid residues of the ACE2 alpha-helix α1, which is necessary for the Spike RBD-ACE2 interface and mobilized during the in vitro viral infection of cells, we demonstrated that the WT ACE2 peptide protects susceptible K18-hACE2 mice against in vivo SARS-CoV-2 infection and is effective for the treatment of COVID-19.
Collapse
Affiliation(s)
- Ernna H Oliveira
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Ana C Monteleone-Cassiano
- Program in Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Lucas Tavares
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Jadson C Santos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Thais M Lima
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Giovanni F Gomes
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Pedro P Tanaka
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Cintia J Monteiro
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Matheus Munuera
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Sabrina S Batah
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Alexandre T Fabro
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Vitor M Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Ana P Masson
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Eduardo A Donadi
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Mariangela Dametto
- Renato Archer Technology Information Center, Ministry of Science, Technology and Innovation, Campinas, SP, Brazil
| | - Rodrigo Bonacin
- Renato Archer Technology Information Center, Ministry of Science, Technology and Innovation, Campinas, SP, Brazil
| | - Ronaldo B Martins
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Eurico Arruda Neto
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Luis Lamberti P daSilva
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Geraldo A Passos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil; Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
23
|
Chikopela T, Mwesigwa N, Masenga SK, Kirabo A, Shibao CA. The Interplay of HIV and Long COVID in Sub-Saharan Africa: Mechanisms of Endothelial Dysfunction. Curr Cardiol Rep 2024; 26:859-871. [PMID: 38958890 DOI: 10.1007/s11886-024-02087-6] [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] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Long COVID affects approximately 5 million people in Africa. This disease is characterized by persistent symptoms or new onset of symptoms after an acute SARS-CoV-2 infection. Specifically, the most common symptoms include a range of cardiovascular problems such as chest pain, orthostatic intolerance, tachycardia, syncope, and uncontrolled hypertension. Importantly, these conditions appear to have endothelial dysfunction as the common denominator, which is often due to impaired nitric oxide (NO) mechanisms. This review discusses the role of mechanisms contributing to endothelial dysfunction in Long COVID, particularly in people living with HIV. RECENT FINDINGS Recent studies have reported that increased inflammation and oxidative stress, frequently observed in Long COVID, may contribute to NO dysfunction, ultimately leading to decreased vascular reactivity. These mechanisms have also been reported in people living with HIV. In regions like Africa, where HIV infection is still a major public health challenge with a prevalence of approximately 26 million people in 2022. Specifically, endothelial dysfunction has been reported as a major mechanism that appears to contribute to cardiovascular diseases and the intersection with Long COVID mechanisms is of particular concern. Further, it is well established that this population is more likely to develop Long COVID following infection with SARS-CoV-2. Therefore, concomitant infection with SARS-CoV-2 may lead to accelerated cardiovascular disease. We outline the details of the worsening health problems caused by Long COVID, which exacerbate pre-existing conditions such as endothelial dysfunction. The overlapping mechanisms of HIV and SARS-CoV-2, particularly the prolonged inflammatory response and chronic hypoxia, may increase susceptibility to Long COVID. Addressing these overlapping health issues is critical as it provides clinical entry points for interventions that could improve and enhance outcomes and quality of life for those affected by both HIV and Long COVID in the region.
Collapse
Affiliation(s)
- Theresa Chikopela
- Department of Human Physiology, Faculty of Medicine, Lusaka Apex Medical University, Lusaka, Zambia
| | - Naome Mwesigwa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Sepiso K Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone, Zambia
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Cyndya A Shibao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA.
| |
Collapse
|
24
|
Luckner KM, Seckel MA. Understanding the Evolving Pathophysiology of Coronavirus Disease 2019 and Adult Nursing Management. Crit Care Nurs Clin North Am 2024; 36:295-321. [PMID: 39069352 DOI: 10.1016/j.cnc.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Coronavirus disease 2019 (COVID-19) was first identified in December 2019 and quickly became a global pandemic. The understanding of the pathophysiology, treatment, and management of the disease has evolved since the beginning of the pandemic in 2020. COVID-19 can be complicated by immune system dysfunction, lung injury with hypoxemia, acute kidney injury, and coagulopathy. The treatment and management of COVID-19 is based on the severity of illness, ranging from asymptomatic to severe and often life-threatening disease. The 3 main recommended medication classes include antivirals, immunomodulators, and anticoagulants. Other supportive therapies include ensuring adequate oxygenation, mechanical ventilation, and prone positioning.
Collapse
|
25
|
Xu W, Langhans SA, Johnson DK, Stauff E, Kandula VVR, Kecskemethy HH, Averill LW, Yue X. Radiotracers for Molecular Imaging of Angiotensin-Converting Enzyme 2. Int J Mol Sci 2024; 25:9419. [PMID: 39273366 PMCID: PMC11395405 DOI: 10.3390/ijms25179419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Angiotensin-converting enzymes (ACE) are well-known for their roles in both blood pressure regulation via the renin-angiotensin system as well as functions in fertility, immunity, hematopoiesis, and many others. The two main isoforms of ACE include ACE and ACE-2 (ACE2). Both isoforms have similar structures and mediate numerous effects on the cardiovascular system. Most remarkably, ACE2 serves as an entry receptor for SARS-CoV-2. Understanding the interaction between the virus and ACE2 is vital to combating the disease and preventing a similar pandemic in the future. Noninvasive imaging techniques such as positron emission tomography and single photon emission computed tomography could noninvasively and quantitatively assess in vivo ACE2 expression levels. ACE2-targeted imaging can be used as a valuable tool to better understand the mechanism of the infection process and the potential roles of ACE2 in homeostasis and related diseases. Together, this information can aid in the identification of potential therapeutic drugs for infectious diseases, cancer, and many ACE2-related diseases. The present review summarized the state-of-the-art radiotracers for ACE2 imaging, including their chemical design, pharmacological properties, radiochemistry, as well as preclinical and human molecular imaging findings. We also discussed the advantages and limitations of the currently developed ACE2-specific radiotracers.
Collapse
Affiliation(s)
- Wenqi Xu
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Sigrid A Langhans
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Division of Neurology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - David K Johnson
- Computational Chemical Biology Core, Molecular Graphics and Modeling Laboratory, University of Kansas, Lawrence, KS 66047, USA
| | - Erik Stauff
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Vinay V R Kandula
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Heidi H Kecskemethy
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Lauren W Averill
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Xuyi Yue
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| |
Collapse
|
26
|
Moon SY, Son M, Kang YW, Koh M, Lee JY, Baek YH. Association between ursodeoxycholic acid use and COVID-19 in individuals with chronic liver disease: a nationwide case-control study in South Korea. Virol J 2024; 21:202. [PMID: 39192342 DOI: 10.1186/s12985-024-02464-1] [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: 05/22/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Conflicting evidence exists regarding the effects of ursodeoxycholic acid (UDCA) on coronavirus disease 2019 (COVID-19). This study investigates the association between UDCA administration and COVID-19 infection and its related outcomes in individuals with chronic liver disease (CLD). METHODS A customized COVID-19 research database (n = 3,485,376) was created by integrating data from the National Health Insurance Service (NHIS) and the Korea Disease Control and Prevention Agency's COVID-19 databases. The study focused on patients diagnosed with COVID-19 in 2021, using the NHIS data from 365 days before diagnosis. To create comparable groups with and without UDCA administration before COVID-19, we used propensity score matching. The primary endpoint was the first confirmed positive result for severe acute respiratory syndrome coronavirus-2. In addition, we identified severe COVID-19-related outcomes. Subgroup analysis were conducted based on the dose of UDCA exposure. RESULTS Data from 74,074 individuals with CLD was analyzed. The participants' average age was 57.5 years, and 52.1% (19,277) of those in each group were male. Those with prior UDCA exposure had a significantly lower risk of COVID-19 infection (adjusted OR: 0.80, 95% CI [0.76-0.85]) compared to the non-UDCA group. Additionally, the UDCA group had a lower risk of severe COVID-19 outcomes (adjusted OR: 0.67, 95% CI [0.46-0.98]). Subgroup analyses indicated that there was a decrease in COVID-19 infection and its related outcomes with increasing UDCA exposure dose. CONCLUSIONS Our large observational study highlights the potential use of readily available UDCA as an adjunctive therapy for COVID-19 in individuals with CLD.
Collapse
Affiliation(s)
- Sang Yi Moon
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea
- Department of Data Sciences Convergence, Dong-A University Interdisciplinary Program, Busan, South Korea
| | - Minkook Son
- Department of Data Sciences Convergence, Dong-A University Interdisciplinary Program, Busan, South Korea
- Department of Physiology, Dong-A University College of Medicine, Busan, South Korea
| | - Yeo Wool Kang
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea
| | - Myeongseok Koh
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea
| | - Jong Yoon Lee
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea
| | - Yang Hyun Baek
- Department of Internal Medicine, Dong-A University College of Medicine, 32 Daesingongwon-ro, Seo-gu, Busan, 49201, South Korea.
| |
Collapse
|
27
|
Ghanem L, Essayli D, Kotaich J, Zein MA, Sahebkar A, Eid AH. Phenotypic switch of vascular smooth muscle cells in COVID-19: Role of cholesterol, calcium, and phosphate. J Cell Physiol 2024:e31424. [PMID: 39188012 DOI: 10.1002/jcp.31424] [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: 03/06/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Although the novel coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), primarily manifests as severe respiratory distress, its impact on the cardiovascular system is also notable. Studies reveal that COVID-19 patients often suffer from certain vascular diseases, partly attributed to increased proliferation or altered phenotype of vascular smooth muscle cells (VSMCs). Although the association between COVID-19 and VSMCs is recognized, the precise mechanism underlying SARS-CoV-2's influence on VSMC phenotype remains largely under-reviewed. In this context, while there is a consistent body of literature dissecting the effect of COVID-19 on the cardiovascular system, few reports delve into the potential role of VSMC switching in the pathophysiology associated with COVID-19 and the molecular mechanisms involved therein. This review dissects and critiques the link between COVID-19 and VSMCs, with particular attention to pathways involving cholesterol, calcium, and phosphate. These pathways underpin the interaction between the virus and VSMCs. Such interaction promotes VSMC proliferation, and eventually potentiates vascular calcification as well as worsens prognosis in patients with COVID-19.
Collapse
Affiliation(s)
- Laura Ghanem
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Dina Essayli
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Jana Kotaich
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
- MEDICA Research Investigation, Beirut, Lebanon
| | - Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
28
|
Wojas-Krawczyk K, Krawczyk P, Błach J, Kucharczyk T, Grenda A, Krzyżanowska N, Szklener K, Horaczyńska-Wojtaś A, Wójcik-Superczyńska M, Chmielewska I, Milanowski J. Immunological insights: assessing immune parameters in medical professionals exposed to SARS-CoV-2. BMC Infect Dis 2024; 24:865. [PMID: 39187767 PMCID: PMC11348584 DOI: 10.1186/s12879-024-09772-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND The immunological background responsible for the severe course of COVID-19 and the immune factors that protect against SARS-CoV-2 infection are still unclear. The aim of this study was to investigate immune system status in persons with high exposure to SARS-CoV-2 infection. METHODS Seventy-one persons employed in the observation and infectious diseases unit were qualified for the study between November 2020 and October 2021. Symptomatic COVID-19 was diagnosed in 35 persons. Anti-SARS-CoV-2 antibodies were also found in 8 persons. Peripheral blood mononuclear cells subpopulations were analyzed by flow cytometry, and the concentrations of cytokines and anti-SARS-CoV-2 antibodies were determined by ELISA. RESULTS The percentages of cytotoxic T lymphocytes (CTLs), CD28+ and T helper (Th) cells with invariant T-cell receptors were significantly higher in persons with symptomatic COVID-19 than in those who did not develop COVID-19' symptoms. Conversely, symptomatic COVID-19 persons had significantly lower percentages of: a) CTLs in the late stage of activation (CD8+/CD95+), b) NK cells, c) regulatory-like Th cells (CD4+/CTLA-4+), and d) Th17-like cells (CD4+/CD161+) compared to asymptomatic COVID-19' persons. Additionally, persons with anti-SARS-CoV-2 antibodies had a significantly higher lymphocyte count and IL-6 concentration than persons without these antibodies. CONCLUSION Numerous lymphocyte populations are permanently altered by SARS-CoV-2 infection. High percentages of both populations: NK cells-as a part of the non-specific response, and T helper cells' as those regulating the immune response, could protect against the acute COVID-19 symptoms development. Understanding the immune background of COVID-19 may improve the prevention of this disease by identifying people at risk of a severe course of infection. TRIAL REGISTRATION This is a retrospective observational study without a trial registration number.
Collapse
Affiliation(s)
- Kamila Wojas-Krawczyk
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland.
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Justyna Błach
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
- Department of Clinical Immunology Medical University of Lublin, Lublin, Poland
| | - Tomasz Kucharczyk
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Anna Grenda
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Natalia Krzyżanowska
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy Medical University of Lublin, Lublin, Poland
| | - Anna Horaczyńska-Wojtaś
- Department of Pediatric Otolaryngology, Phoniatrics and Audiology, University Children's Hospital, Lublin, Poland
| | - Magdalena Wójcik-Superczyńska
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Izabela Chmielewska
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology Medical University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| |
Collapse
|
29
|
Cuevas-Sierra A, Chero-Sandoval L, Higuera-Gómez A, Vargas JA, Martínez-Urbistondo M, Castejón R, Martínez JA. Modulatory role of Faecalibacterium on insulin resistance and coagulation in patients with post-viral long haulers depending on adiposity. iScience 2024; 27:110450. [PMID: 39081294 PMCID: PMC11284562 DOI: 10.1016/j.isci.2024.110450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/05/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Patients with Post-viral long hauler encompass lasting symptoms and comorbid complexities, often exacerbated in individuals with excessive body weight. The aim was to study gut microbiota in 130 patients with post-viral long hauler stratified by body mass index (BMI) and the relationship between inflammation and microbiota. Significant higher values were found for anthropometric variables and markers of glucose and dyslipidemia in individuals with higher BMI, as well as elevated levels of C-reactive protein, fibrinogen, IL-6, uric acid, and D-dimer. An interactive association showed an interplay between Faecalibacterium, D-dimer levels, and insulin resistance. This investigation showed that anthropometric, biochemical, and inflammatory variables were impaired in patients with post-viral long haulers with higher BMI. In addition, gut microbiota differences were found between groups and a modification effect on Faecalibacterium abundance regarding insulin resistance and D-dimer. These findings suggest that considering adiposity and gut microbiota structure and composition may improve personalized clinical interventions in patients with chronic inflammation.
Collapse
Affiliation(s)
- Amanda Cuevas-Sierra
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - Lourdes Chero-Sandoval
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
- Department of Endocrinology and Nutrition of the University Clinical Hospital, University of Valladolid, 47002 Valladolid, Spain
| | - Andrea Higuera-Gómez
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
| | - J. Antonio Vargas
- Internal Medicine Service of Puerta de Hierro Majadahonda University Hospital, 2822 Madrid, Spain
| | | | - Raquel Castejón
- Internal Medicine Service of Puerta de Hierro Majadahonda University Hospital, 2822 Madrid, Spain
| | - J. Alfredo Martínez
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain
- Centro de Medicina y Endocrinología, Universidad de Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
30
|
Lopez L, Kogut K, Rauch S, Gunier RB, Wong MP, Harris E, Deardorff J, Eskenazi B, Harley KG. Organophosphate pesticide exposure and risk of SARS-CoV-2 infection. ENVIRONMENTAL RESEARCH 2024; 255:119214. [PMID: 38788790 DOI: 10.1016/j.envres.2024.119214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Several studies have reported immune modulation by organophosphate (OP) pesticides, but the relationship between OP exposure and SARS-CoV-2 infection is yet to be studied. We used two different measures of OP pesticide exposure (urinary biomarkers (N = 154) and residential proximity to OP applications (N = 292)) to examine the association of early-childhood and lifetime exposure to OPs and risk of infection of SARS-CoV-2 using antibody data. Our study population consisted of young adults (ages 18-21 years) from the Center for the Health Assessment of Mothers and Children of Salinas (CHAMACOS) Study, a longitudinal cohort of families from a California agricultural region. Urinary biomarkers reflected exposure from in utero to age 5 years. Residential proximity reflected exposures between in utero and age 16 years. SARS-CoV-2 antibodies in blood samples collected between June 2022 and January 2023 were detected via two enzyme linked immunosorbent assays, each designed to bind to different SARS-CoV-2 antigens. We performed logistic regression for each measure of pesticide exposure, adjusting for covariates from demographic data and self-reported questionnaire data. We found increased odds of SARS-CoV-2 infection among participants with higher urinary biomarkers of OPs in utero (OR = 1.94, 95% CI: 0.71, 5,58) and from age 0-5 (OR = 1.90, 95% CI: 0.54, 6.95).
Collapse
Affiliation(s)
- Luis Lopez
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, United States
| | - Katie Kogut
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California, Berkeley, Berkeley, United States
| | - Stephen Rauch
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California, Berkeley, Berkeley, United States
| | - Robert B Gunier
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California, Berkeley, Berkeley, United States
| | - Marcus P Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, United States
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, United States
| | - Julianna Deardorff
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California, Berkeley, Berkeley, United States
| | - Brenda Eskenazi
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California, Berkeley, Berkeley, United States
| | - Kim G Harley
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California, Berkeley, Berkeley, United States.
| |
Collapse
|
31
|
Eliadis P, Mais A, Papazisis A, Loxa EK, Dimitriadis A, Sarrigeorgiou I, Backovic M, Agallou M, Zouridakis M, Karagouni E, Lazaridis K, Mamalaki A, Lymberi P. Novel Competitive ELISA Utilizing Trimeric Spike Protein of SARS-CoV-2, Could Identify More Than RBD-RBM Specific Neutralizing Antibodies in Hybrid Sera. Vaccines (Basel) 2024; 12:914. [PMID: 39204038 PMCID: PMC11359269 DOI: 10.3390/vaccines12080914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Since the initiation of the COVID-19 pandemic, there has been a need for the development of diagnostic methods to determine the factors implicated in mounting an immune response against the virus. The most promising indicator has been suggested to be neutralizing antibodies (nAbs), which mainly block the interaction between the Spike protein (S) of SARS-CoV-2 and the host entry receptor ACE2. In this study, we aimed to develop and optimize conditions of a competitive ELISA to measure serum neutralizing titer, using a recombinant trimeric Spike protein modified to have six additional proline residues (S(6P)-HexaPro) and h-ACE2. The results of our surrogate Virus Neutralizing Assay (sVNA) were compared against the commercial sVNT (cPass, Nanjing GenScript Biotech Co., Nanjing City, China), using serially diluted sera from vaccinees, and a high correlation of ID50-90 titer values was observed between the two assays. Interestingly, when we tested and compared the neutralizing activity of sera from eleven fully vaccinated individuals who subsequently contracted COVID-19 (hybrid sera), we recorded a moderate correlation between the two assays, while higher sera neutralizing titers were measured with sVNA. Our data indicated that the sVNA, as a more biologically relevant model assay that paired the trimeric S(6P) with ACE2, instead of the isolated RBD-ACE2 pairing cPass test, could identify nAbs other than the RBD-RBM specific ones.
Collapse
Affiliation(s)
- Petros Eliadis
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
- Biotechnology Unit, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.D.); (A.M.)
| | - Annie Mais
- Laboratory of Molecular Biology and Immunobiotechnology, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Alexandros Papazisis
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
| | - Eleni K. Loxa
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
| | - Alexios Dimitriadis
- Biotechnology Unit, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.D.); (A.M.)
| | - Ioannis Sarrigeorgiou
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
| | - Marija Backovic
- Institut Pasteur, Unité de Virologie Structurale, Université Paris Cité, CNRS-UMR3569, 75724 Paris, France;
| | - Maria Agallou
- Immunology of Infection Laboratory, Microbiology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (M.A.); (E.K.)
| | - Marios Zouridakis
- Structural Neurobiology Research Group, Laboratory of Molecular Neurobiology and Immunology, Department of Neurobiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Evdokia Karagouni
- Immunology of Infection Laboratory, Microbiology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (M.A.); (E.K.)
| | - Konstantinos Lazaridis
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
- Biotechnology Unit, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.D.); (A.M.)
| | - Avgi Mamalaki
- Biotechnology Unit, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.D.); (A.M.)
- Laboratory of Molecular Biology and Immunobiotechnology, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Peggy Lymberi
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, 11521 Athens, Greece; (A.P.); (E.K.L.); (I.S.); (K.L.)
| |
Collapse
|
32
|
Ma H, Hao J, Li W, Yu K, Zhu K, Yang M, Cao S, Xue H, Liu D, Song Y, Zhang S, Zhang X, Sun Z, Gao X. Evaluation of feline mesenchymal stem cell susceptibility to feline viruses. Sci Rep 2024; 14:18598. [PMID: 39127765 PMCID: PMC11316800 DOI: 10.1038/s41598-024-69343-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024] Open
Abstract
Feline mesenchymal stem cells (fMSCs) are well known for their robust differentiation capabilities and are commonly used in studying immune-related diseases in cats. Despite their importance, the susceptibility of fMSCs to viral infections remains uncertain. This study aimed to assess the susceptibility of feline adipose-derived mesenchymal stem cells (fAD-MSCs) and feline umbilical cord-derived mesenchymal stem cells (fUC-MSCs) to common feline viruses, including feline coronavirus (FCoV), feline herpesvirus type 1 (FHV-1), and feline panleukopenia virus (FPV). The results demonstrated that both FCoV and FHV-1 were able to infect both types of cells, while FPV did not exhibit cytopathic effects on fUC-MSCs. Furthermore, all three viruses were successfully isolated from fAD-MSCs. These findings suggest that certain feline viruses can replicate in fMSCs, indicating potential limitations in using fMSCs for treating viral diseases caused by these specific viruses. This study has important clinical implications for veterinarians, particularly in the management of viral diseases.
Collapse
Affiliation(s)
- Haoyuan Ma
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Jingrui Hao
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Weijian Li
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Kai Yu
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Kunru Zhu
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Meng Yang
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Shuoning Cao
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Haowen Xue
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Dan Liu
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Yanhao Song
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Siqi Zhang
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Xifeng Zhang
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Zheng Sun
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China
| | - Xu Gao
- Laboratory for Animal Molecular Virology, Department of Veterinary Medicine, College of Agricultural, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
33
|
Xiang Y, Feng Y, Qiu J, Zhang R, So HC. Association of COVID-19 vaccination with risks of hospitalization due to cardiovascular and other diseases: A study using data from the UK Biobank. Int J Infect Dis 2024; 145:107080. [PMID: 38701913 DOI: 10.1016/j.ijid.2024.107080] [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: 01/23/2024] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVES To explore whether COVID-19 vaccination protects against hospital admission by preventing infections and severe disease. METHODS We leveraged the UK Biobank and studied associations of COVID-19 vaccination (BioNTech-BNT162b2 or Oxford-AstraZeneca-ChAdOx1) with hospitalizations from cardiovascular and other selected diseases (N = 393,544; median follow-up = 54 days among vaccinated individuals). Multivariable Cox, Poisson regression, propensity score matching, and inverse probability treatment weighting analyses were performed. We also performed adjustment using prescription-time distribution matching, and prior event rate ratio. RESULTS We observed that COVID-19 vaccination (at least one dose), compared with no vaccination, was associated with reduced short-term risks of hospitalizations from stroke (hazard ratio [HR] = 0.178, 95% confidence interval [CI]: 0.127-0.250, P = 1.50e-23), venous thromboembolism (HR = 0.426, CI: 0.270-0.673, P = 2.51e-4), dementia (HR = 0.114, CI: 0.060-0.216; P = 2.24e-11), non-COVID-19 pneumonia (HR = 0.108, CI: 0.080-0.145; P = 2.20e-49), coronary artery disease (HR = 0.563, CI: 0.416-0.762; P = 2.05e-4), chronic obstructive pulmonary disease (HR = 0.212, CI: 0.126-0.357; P = 4.92e-9), type 2 diabetes (HR = 0.216, CI: 0.096-0.486, P = 2.12e-4), heart failure (HR = 0.174, CI: 0.118-0.256, P = 1.34e-18), and renal failure (HR = 0.415, CI: 0.255-0.677, P = 4.19e-4), based on standard Cox regression models. Among the previously mentioned results, reduced hospitalizations for stroke, heart failure, non-COVID-19 pneumonia, and dementia were consistently observed across regression, propensity score matching/inverse probability treatment weighting, prescription-time distribution matching, and prior event rate ratio. The results for two-dose vaccination were similar. CONCLUSIONS Taken together, this study provides further support to the safety and benefits of COVID-19 vaccination, and such benefits may extend beyond reduction of infection risk or severity per se. However, causal relationship cannot be concluded and further studies are required.
Collapse
Affiliation(s)
- Yong Xiang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yaning Feng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinghong Qiu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ruoyu Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology and The Chinese University of Hong Kong, Hong Kong, China; CUHK Shenzhen Research Institute, Shenzhen, China; Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong; Margaret K.L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong; Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, Hong Kong; Hong Kong Branch of the Chinese Academy of Sciences Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
34
|
Gheban-Roşca IA, Gheban BA, Pop B, Mironescu DC, Siserman VC, Jianu EM, Drugan T, Bolboacă SD. A histopathological analysis of extrapulmonary lesions in fatal coronavirus disease (COVID-19). Pathol Res Pract 2024; 260:155373. [PMID: 38901140 DOI: 10.1016/j.prp.2024.155373] [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: 02/06/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
INTRODUCTION The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents diverse clinical manifestations and multi-organ involvement. This study aimed to evaluate the extra-pulmonary histopathological patterns underpinning COVID-19-induced lesions in cardiac, hepatic, renal, brainstem, and splenic tissues. MATERIALS AND METHODS The research involved conventional forensic autopsies conducted between April 2020 and April 2021 on individuals with confirmed SARS-CoV-2 infection in Cluj-Napoca, Romania. Tissues were processed and stained for histological examination. Differences in patients with and without diffuse alveolar damage (DAD) were evaluated. RESULTS In our study of 79 COVID-19 autopsies conducted on unvaccinated patients besides lung involvement, the patients had histological changes in at least two out of five (brain, heart, liver, kidney, and spleen) organs. Notable findings include hepatitis observed in 46.8 % of cases, 21.5 % with lobular hepatitis, and 41.8 % with liver steatosis. Additionally, 69.6 % exhibited acute tubular necrosis, and 55.7 % had varying degrees of splenic lymphocyte depletion. Almost 41 % of cases had pericardial effusion, 36.7 % myocarditis, 24.1 % myocardial infarction, and 12.7 % of cases had encephalitis. Acute tubular necrosis (78.6 %) was the most frequent histopathological finding observed in patients with DAD. Myocarditis was described in 45.9 % of the patients without DAD. DISCUSSION The autopsy findings in our cohort of COVID-19 victims align with international scientific literature. Distinguishing viral-induced myocarditis, encephalitis, hepatitis, or systemic inflammatory syndrome remains challenging. CONCLUSION Post-mortem analysis identified lesions associated with SARS-CoV-2 in multiple organs, highlighting the systemic nature of the virus and emphasizing the need for continued research into organ-specific damage and long-term sequelae of COVID-19.
Collapse
Affiliation(s)
- Ioana-Andreea Gheban-Roşca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania; Clinical Hospital for Infectious Diseases, Cluj-Napoca 400003, Romania
| | - Bogdan-Alexandru Gheban
- County Emergency Clinical Hospital, Cluj-Napoca 400347, Romania; Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania.
| | - Bogdan Pop
- The Oncology Institute " Prof. Dr. Ion Chiricuță", Cluj-Napoca 400015, Romania; Department of Anatomic Pathology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Daniela-Cristina Mironescu
- Forensic Institute, Cluj-Napoca 400006, Romania; Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Vasile Costel Siserman
- Forensic Institute, Cluj-Napoca 400006, Romania; Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Elena Mihaela Jianu
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Tudor Drugan
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Sorana D Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| |
Collapse
|
35
|
Holodinsky JK, Kumar M, McNaughton CD, Austin PC, Chu A, Hill MD, Norris C, Field TS, Lee DS, Kapral MK, Kamal N, Yu AY. An Interrupted Time-Series Analysis of the Impact of COVID-19 on Hospitalizations for Vascular Events in 3 Canadian Provinces. CJC Open 2024; 6:959-966. [PMID: 39211760 PMCID: PMC11357754 DOI: 10.1016/j.cjco.2024.04.010] [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: 04/05/2024] [Accepted: 04/26/2024] [Indexed: 09/04/2024] Open
Abstract
Background COVID-19 infection is associated with a pro-coagulable state, thrombosis, and cardiovascular events. However, its impact on population-based rates of vascular events is less well understood. We studied temporal trends in hospitalizations for stroke and myocardial infarction in 3 Canadian provinces (Alberta, Ontario, and Nova Scotia) between 2014 and 2022. Methods Linked administrative data from each province were used to identify admissions for ischemic stroke, intracerebral hemorrhage, cerebral venous thrombosis, and myocardial infarction. Event rates per 100,000/quarter, standardized to the 2016 Canadian population, were calculated. We assessed changes from quarterly rates pre-pandemic (2014-2020), compared to rates in the pandemic period (2020-2022), using interrupted time-series analysis with a jump discontinuity at pandemic onset. Age group- and sex-stratified analyses also were performed. Results We identified 162,497 strokes and 243,182 myocardial infarctions. At pandemic onset, no significant step change in strokesper 100,000/quarter was observed in any of the 3 provinces. During the pandemic, stroke rates were stable in Alberta and Ontario, but they increased in Nova Scotia (0.44 per 100,000/quarter, P = 0.004). At pandemic onset, a significant step decrease occurred in myocardial infarctions per 100,000/quarter in Alberta (4.72, P < 0.001) and Ontario (4.84, P < 0.001), but not in Nova Scotia. During the pandemic, myocardial infarctions per 100,000/quarter decreased in Alberta (-0.34, P = 0.01), but they remained stable in Ontario and Nova Scotia. No consistent patterns by age group or sex were noted. Conclusions Hospitalization rates for stroke or myocardial infarction across 3 Canadian provinces did not increase substantially during the first 2 years of the pandemic. Continued surveillance is warranted as the virus becomes endemic.
Collapse
Affiliation(s)
- Jessalyn K. Holodinsky
- Department of Emergency Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Center for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O’Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mukesh Kumar
- Department of Industrial Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Candace D. McNaughton
- ICES, Toronto, Ontario, Canada
- Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Michael D. Hill
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Colleen Norris
- Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada
| | - Thalia S. Field
- Vancouver Stroke Program, Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Douglas S. Lee
- ICES, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Moira K. Kapral
- ICES, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine (General Internal Medicine), University of Toronto, Toronto, Ontario, Canada
| | - Noreen Kamal
- Department of Industrial Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Community Health and Epidemiology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine (Neurology), Dalhousie University, Halifax, Nova Scotia, Canada
| | - Amy Y.X. Yu
- ICES, Toronto, Ontario, Canada
- Department of Medicine (Neurology), University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Loo C, Treacy MG, Toerien L, Msellati A, Catanzano T. Emergency Presentations of Coronavirus Disease (COVID-19): A Review of the Literature and Radiologic Perspective. Semin Ultrasound CT MR 2024; 45:332-338. [PMID: 38996944 DOI: 10.1053/j.sult.2024.07.003] [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: 07/14/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the debilitating global pandemic known as Coronavirus disease (COVID-19). In this paper, we highlight the widespread manifestations and complications across disease systems. In addition, we present their relevant imaging findings to inform appropriate investigations and management in patients presenting to the Emergency Department with COVID-19 and its respective sequalae. Of note, we outline considerations for diagnosis of long COVID, an important medium to long term sequalae in patients with previous COVID-19 infections.
Collapse
Affiliation(s)
- Caitlyn Loo
- School of Medicine, University College Dublin, Belfield, Ireland; Department of Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Molly Godson Treacy
- Department of Radiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Lara Toerien
- Department of Radiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | | | - Tara Catanzano
- Department of Radiology, Baystate Health, Springfield, MA.
| |
Collapse
|
37
|
Hotowy A, Strojny-Cieślak B, Ostrowska A, Zielińska-Górska M, Kutwin M, Wierzbicki M, Sosnowska M, Jaworski S, Chwalibóg A, Kotela I, Sawosz Chwalibóg E. Silver and Carbon Nanomaterials/Nanocomplexes as Safe and Effective ACE2-S Binding Blockers on Human Skin Cell Lines. Molecules 2024; 29:3581. [PMID: 39124987 PMCID: PMC11313757 DOI: 10.3390/molecules29153581] [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: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
(1) Background: Angiotensin-converting enzyme 2 (ACE2) is a crucial functional receptor of the SARS-CoV-2 virus. Although the scale of infections is no longer at pandemic levels, there are still fatal cases. The potential of the virus to infect the skin raises questions about new preventive measures. In the context of anti-SARS-CoV-2 applications, the interactions of antimicrobial nanomaterials (silver, Ag; diamond, D; graphene oxide, GO and their complexes) were examined to assess their ability to affect whether ACE2 binds with the virus. (2) Methods: ACE2 inhibition competitive tests and in vitro treatments of primary human adult epidermal keratinocytes (HEKa) and primary human adult dermal fibroblasts (HDFa) were performed to assess the blocking capacity of nanomaterials/nanocomplexes and their toxicity to cells. (3) Results: The nanocomplexes exerted a synergistic effect compared to individual nanomaterials. HEKa cells were more sensitive than HDFa cells to Ag treatments and high concentrations of GO. Cytotoxic effects were not observed with D. In the complexes, both carbonic nanomaterials had a soothing effect against Ag. (4) Conclusions: The Ag5D10 and Ag5GO10 nanocomplexes seem to be most effective and safe for skin applications to combat SARS-CoV-2 infection by blocking ACE2-S binding. These nanocomplexes should be evaluated through prolonged in vivo exposure. The expected low specificity enables wider applications.
Collapse
Affiliation(s)
- Anna Hotowy
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| | - Barbara Strojny-Cieślak
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| | - Agnieszka Ostrowska
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| | - Marlena Zielińska-Górska
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| | - Marta Kutwin
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| | - Mateusz Wierzbicki
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| | - Malwina Sosnowska
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| | - André Chwalibóg
- Section of Production, Nutrition and Health, Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1870 Frederiksberg, Denmark
| | - Ireneusz Kotela
- Department of Orthopaedics, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland;
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-369 Kielce, Poland
| | - Ewa Sawosz Chwalibóg
- Department of Nanobiotechnology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (B.S.-C.); (A.O.); (M.Z.-G.); (M.K.); (M.W.); (M.S.); (S.J.); (E.S.C.)
| |
Collapse
|
38
|
Guarienti FA, Xavier FAC, Ferraz MD, Wagner F, Marinowic DR, da Costa JC, Machado DC. Are COVID-19 Polymorphisms in ACE and ACE2 Prognosis Predictors? Curr Issues Mol Biol 2024; 46:8111-8117. [PMID: 39194697 DOI: 10.3390/cimb46080480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 08/29/2024] Open
Abstract
Regardless of the containment of the SARS-CoV-2 pandemic, it remains paramount to comprehensively understand its underlying mechanisms to mitigate potential future health and economic impacts, comparable to those experienced throughout the course of the pandemic. The angiotensin-converting enzyme 2 (ACE2) provides anchorage for SARS-CoV-2 binding, thus implicating that ACE and ACE2 might contribute to the variability in infection severity. This study aimed to elucidate predisposing factors influencing the disease course among people infected by SARS-CoV-2, focusing on angiotensin-converting enzyme (ACE) and ACE2 polymorphisms. Notably, despite similar demographics and comorbidities, COVID-19 patients exhibit substantial differences in prognosis. Genetic polymorphisms in ACE and ACE2 have been implicated in disease progression, prompting our investigation into their role in COVID-19 evolution. Using next-generation sequencing (NGS), we analyzed ACE and ACE2 genes in a sample group comprising six subjects infected by SARS-CoV-2. Our findings revealed a correlation between specific polymorphisms and COVID-19 outcomes. Specifically, ACE and ACE2 intronic deletions were observed in all deceased patients, suggesting a potential association with mortality. These results highlight the significance of genetic factors in shaping the clinical course of COVID-19, emphasizing the importance of further research into the impact of genetic variations on COVID-19 severity.
Collapse
Affiliation(s)
- Fabiana Amaral Guarienti
- Post Graduation Program of Geriatrics and Gerontology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
| | - Fernando Antônio Costa Xavier
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Precision Medicine and Biotechnology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
| | - Mateus Duarte Ferraz
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
| | - Fernanda Wagner
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Precision Medicine and Biotechnology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
| | - Daniel Rodrigo Marinowic
- Post Graduation Program of Geriatrics and Gerontology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Precision Medicine and Biotechnology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
| | - Jaderson Costa da Costa
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Neurosciences and Electrophisiology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
| | - Denise Cantarelli Machado
- Post Graduation Program of Geriatrics and Gerontology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
- Laboratory of Molecular Biology and Immunology, Brain Institute of Rio Grande do Sul, Porto Alegre 90610, Brazil
- Post Graduation Program of Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619, Brazil
| |
Collapse
|
39
|
Takeda Y, Demura M, Yoneda T, Takeda Y. Epigenetic Regulation of the Renin-Angiotensin-Aldosterone System in Hypertension. Int J Mol Sci 2024; 25:8099. [PMID: 39125667 PMCID: PMC11312206 DOI: 10.3390/ijms25158099] [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: 05/29/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Activation of the renin-angiotensin-aldosterone system (RAAS) plays an important pathophysiological role in hypertension. Increased mRNA levels of the angiotensinogen angiotensin-converting enzyme, angiotensin type 1 receptor gene, Agtr1a, and the aldosterone synthase gene, CYP11B2, have been reported in the heart, blood vessels, and kidneys in salt-sensitive hypertension. However, the mechanism of gene regulation in each component of the RAAS in cardiovascular and renal tissues is unclear. Epigenetic mechanisms, which are important for regulating gene expression, include DNA methylation, histone post-translational modifications, and microRNA (miRNA) regulation. A close association exists between low DNA methylation at CEBP-binding sites and increased AGT expression in visceral adipose tissue and the heart of salt-sensitive hypertensive rats. Several miRNAs influence AGT expression and are associated with cardiovascular diseases. Expression of both ACE and ACE2 genes is regulated by DNA methylation, histone modifications, and miRNAs. Expression of both angiotensinogen and CYP11B2 is reversibly regulated by epigenetic modifications and is related to salt-sensitive hypertension. The mineralocorticoid receptor (MR) exists in cardiovascular and renal tissues, in which many miRNAs influence expression and contribute to the pathogenesis of hypertension. Expression of the 11beta-hydroxysteroid dehydrogenase type 2 (HSD11B2) gene is also regulated by methylation and miRNAs. Epigenetic regulation of renal and vascular HSD11B2 is an important pathogenetic mechanism for salt-sensitive hypertension.
Collapse
Affiliation(s)
- Yoshimichi Takeda
- Endocrinology and Metabolism, Saiseikai Kanazawa Hospital, Kanazawa 920-0353, Japan;
- Department of Hygiene, Graduate School of Medical Science, Kanazawa University, Kanazawa 921-8641, Japan;
| | - Masashi Demura
- Department of Hygiene, Graduate School of Medical Science, Kanazawa University, Kanazawa 921-8641, Japan;
| | - Takashi Yoneda
- Institute of Liberal Arts and Science, Kanazawa University, Kanazawa 921-8641, Japan;
- Department of Health Promotion of Medicine of the Future, Graduate School of Medical Science, Kanazawa University, Kanazawa 921-8641, Japan
| | - Yoshiyu Takeda
- Department of Health Promotion of Medicine of the Future, Graduate School of Medical Science, Kanazawa University, Kanazawa 921-8641, Japan
- Hypertension Center, Asanogawa General Hospital, Kanazawa 910-8621, Japan
| |
Collapse
|
40
|
Wijerathne SVT, Pandit R, Ipinmoroti AO, Crenshaw BJ, Matthews QL. Feline coronavirus influences the biogenesis and composition of extracellular vesicles derived from CRFK cells. Front Vet Sci 2024; 11:1388438. [PMID: 39091390 PMCID: PMC11292801 DOI: 10.3389/fvets.2024.1388438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction Coronavirus (CoV) has become a public health crisis that causes numerous illnesses in humans and certain animals. Studies have identified the small, lipid-bound structures called extracellular vesicles (EVs) as the mechanism through which viruses can enter host cells, spread, and evade the host's immune defenses. EVs are able to package and carry numerous viral compounds, including proteins, genetic substances, lipids, and receptor proteins. We proposed that the coronavirus could alter EV production and content, as well as influence EV biogenesis and composition in host cells. Methods In the current research, Crandell-Rees feline kidney (CRFK) cells were infected with feline coronavirus (FCoV) in an exosome-free media at a multiplicity of infection (MOI) of 2,500 infectious units (IFU) at 48 h and 72 h time points. Cell viability was analyzed and found to be significantly decreased by 9% (48 h) and 15% (72 h) due to FCoV infection. EVs were isolated by ultracentrifugation, and the surface morphology of isolated EVs was analyzed via Scanning Electron Microscope (SEM). Results NanoSight particle tracking analysis (NTA) confirmed that the mean particle sizes of control EVs were 131.9 nm and 126.6 nm, while FCoV infected-derived EVs were 143.4 nm and 120.9 nm at 48 and 72 h, respectively. Total DNA, RNA, and protein levels were determined in isolated EVs at both incubation time points; however, total protein was significantly increased at 48 h. Expression of specific protein markers such as TMPRSS2, ACE2, Alix, TSG101, CDs (29, 47, 63), TLRs (3, 6, 7), TNF-α, and others were altered in infection-derived EVs when compared to control-derived EVs after FCoV infection. Discussion Our findings suggested that FCoV infection could alter the EV production and composition in host cells, which affects the infection progression and disease evolution. One purpose of studying EVs in various animal coronaviruses that are in close contact with humans is to provide significant information about disease development, transmission, and adaptation. Hence, this study suggests that EVs could provide diagnostic and therapeutic applications in animal CoVs, and such understanding could provide information to prevent future coronavirus outbreaks.
Collapse
Affiliation(s)
| | - Rachana Pandit
- Microbiology Program, Alabama State University, Montgomery, AL, United States
| | | | | | - Qiana L. Matthews
- Microbiology Program, Alabama State University, Montgomery, AL, United States
- Department of Biological Sciences, College of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, AL, United States
| |
Collapse
|
41
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. Int J Mol Sci 2024; 25:7553. [PMID: 39062796 PMCID: PMC11277036 DOI: 10.3390/ijms25147553] [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: 05/27/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Proteases are produced and released in the mucosal cells of the respiratory tract and have important physiological functions, for example, maintaining airway humidification to allow proper gas exchange. The infectious mechanism of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), takes advantage of host proteases in two ways: to change the spatial conformation of the spike (S) protein via endoproteolysis (e.g., transmembrane serine protease type 2 (TMPRSS2)) and as a target to anchor to epithelial cells (e.g., angiotensin-converting enzyme 2 (ACE2)). This infectious process leads to an imbalance in the mucosa between the release and action of proteases versus regulation by anti-proteases, which contributes to the exacerbation of the inflammatory and prothrombotic response in COVID-19. In this article, we describe the most important proteases that are affected in COVID-19, and how their overactivation affects the three main physiological systems in which they participate: the complement system and the kinin-kallikrein system (KKS), which both form part of the contact system of innate immunity, and the renin-angiotensin-aldosterone system (RAAS). We aim to elucidate the pathophysiological bases of COVID-19 in the context of the imbalance between the action of proteases and anti-proteases to understand the mechanism of aprotinin action (a panprotease inhibitor). In a second-part review, titled "Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions", we explain in depth the pharmacodynamics, pharmacokinetics, toxicity, and use of aprotinin as an antiviral drug.
Collapse
Affiliation(s)
- Juan Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
42
|
Borowczak J, Gąsiorek-Kwiatkowska A, Szczerbowski K, Maniewski M, Zdrenka M, Szadurska-Noga M, Gostomczyk K, Rutkiewicz P, Olejnik K, Cnota W, Karpów-Greiner M, Knypiński W, Sekielska-Domanowska M, Ludwikowski G, Dubiel M, Szylberg Ł, Bodnar M. SARS-CoV-2 Infection during Delivery Causes Histopathological Changes in the Placenta. Diseases 2024; 12:142. [PMID: 39057113 PMCID: PMC11276080 DOI: 10.3390/diseases12070142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND SARS-CoV-2 can damage human placentas, leading to pregnancy complications, such as preeclampsia and premature birth. This study investigates the histopathological changes found in COVID-19-affected placentas. MATERIALS AND METHODS This study included 23 placentas from patients with active COVID-19 during delivery and 22 samples from patients without COVID-19 infection in their medical history. The samples underwent histopathological examination for pathology, such as trophoblast necrosis, signs of vessel damage, or fetal vascular malperfusion. RESULTS Newborns from the research group have lower weights and Apgar scores than healthy newborns. In the COVID-19 group, calcifications and collapsed intervillous space were more frequent, and inflammation was more severe than in the healthy group. At the same time, the placenta of SARS-CoV-2-positive patients showed signs of accelerated vascular maturation. Trophoblast necrosis was found only in the placentas of the research group. The expression of CD68+ was elevated in the COVID-19 cohort, suggesting that macrophages constituted a significant part of the inflammatory infiltrate. The increase in lymphocyte B markers was associated with placental infarctions, while high levels of CD3+, specific for cytotoxic T lymphocytes, correlated with vascular injury. CONCLUSIONS SARS-CoV-2 is associated with pathological changes in the placenta, including trophoblast necrosis, calcification, and accelerated villous maturation. Those changes appear to be driven by T cells and macrophages, whose increased expression reflects ongoing histiocytic intervillositis in the placenta.
Collapse
Affiliation(s)
- Jędrzej Borowczak
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Agnieszka Gąsiorek-Kwiatkowska
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Krzysztof Szczerbowski
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Mateusz Maniewski
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Doctoral School of Medical and Health Sciences, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland
| | - Marek Zdrenka
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
| | - Marta Szadurska-Noga
- Department of Pathomorphology and Forensic Medicine, Faculty of Medical Sciences, University of Warmia and Mazury, 10-561 Olsztyn, Poland;
| | - Karol Gostomczyk
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Paula Rutkiewicz
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Katarzyna Olejnik
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Wojciech Cnota
- Chair and Department of Gynaecology and Obstetrics, Faculty of Health Sciences in Katowice, Medical University of Silesia, 41-703 Ruda Śląska, Poland
| | - Magdalena Karpów-Greiner
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Wojciech Knypiński
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Marta Sekielska-Domanowska
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Grzegorz Ludwikowski
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Mariusz Dubiel
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
| | - Łukasz Szylberg
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland (K.G.)
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| | - Magdalena Bodnar
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-168 Bydgoszcz, Poland (M.K.-G.); (W.K.); (M.S.-D.); (M.D.)
- Chair of Pathology, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland (K.O.)
| |
Collapse
|
43
|
Payen SH, Adhikari K, Petereit J, Uppal T, Rossetto CC, Verma SC. SARS-CoV-2 superinfection in CD14 + monocytes with latent human cytomegalovirus (HCMV) promotes inflammatory cascade. Virus Res 2024; 345:199375. [PMID: 38642618 PMCID: PMC11061749 DOI: 10.1016/j.virusres.2024.199375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of coronavirus disease 2019 (COVID-19), has posed significant challenges to global health. While much attention has been directed towards understanding the primary mechanisms of SARS-CoV-2 infection, emerging evidence suggests co-infections or superinfections with other viruses may contribute to increased morbidity and mortality, particularly in severe cases of COVID-19. Among viruses that have been reported in patients with SARS-CoV-2, seropositivity for Human cytomegalovirus (HCMV) is associated with increased COVID-19 risk and hospitalization. HCMV is a ubiquitous beta-herpesvirus with a seroprevalence of 60-90 % worldwide and one of the leading causes of mortality in immunocompromised individuals. The primary sites of latency for HCMV include CD14+ monocytes and CD34+ hematopoietic cells. In this study, we sought to investigate SARS-CoV-2 infection of CD14+ monocytes latently infected with HCMV. We demonstrate that CD14+ cells are susceptible and permissive to SARS-CoV-2 infection and detect subgenomic transcripts indicative of replication. To further investigate the molecular changes triggered by SARS-CoV-2 infection in HCMV-latent CD14+ monocytes, we conducted RNA sequencing coupled with bioinformatic differential gene analysis. The results revealed significant differences in cytokine-cytokine receptor interactions and inflammatory pathways in cells superinfected with replication-competent SARS-CoV-2 compared to the heat-inactivated and mock controls. Notably, there was a significant upregulation in transcripts associated with pro-inflammatory response factors and a decrease in anti-inflammatory factors. Taken together, these findings provide a basis for the heightened inflammatory response, offering potential avenues for targeted therapeutic interventions among HCMV-infected severe cases of COVID-19. SUMMARY: COVID-19 patients infected with secondary viruses have been associated with a higher prevalence of severe symptoms. Individuals seropositive for human cytomegalovirus (HCMV) infection are at an increased risk for severe COVID-19 disease and hospitalization. HCMV reactivation has been reported in severe COVID-19 cases with respiratory failure and could be the result of co-infection with SARS-CoV-2 and HCMV. In a cell culture model of superinfection, HCMV has previously been shown to increase infection of SARS-CoV-2 of epithelial cells by upregulating the human angiotensin-converting enzyme-2 (ACE2) receptor. In this study, we utilize CD14+ monocytes, a major cell type that harbors latent HCMV, to investigate co-infection of SARS-CoV-2 and HCMV. This study is a first step toward understanding the mechanism that may facilitate increased COVID-19 disease severity in patients infected with SARS-CoV-2 and HCMV.
Collapse
Affiliation(s)
- Shannon Harger Payen
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Kabita Adhikari
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Juli Petereit
- Nevada Bioinformatics Center (RRID:SCR_017802), University of Nevada, Reno, NV 89557, United States
| | - Timsy Uppal
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Cyprian C Rossetto
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Subhash C Verma
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States.
| |
Collapse
|
44
|
Mannheim J, Johnson D. COVID-19 and Diabetes: An Epidemiologic Overview. Pediatr Ann 2024; 53:e258-e263. [PMID: 38949874 DOI: 10.3928/19382359-20240502-07] [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] [Indexed: 07/03/2024]
Abstract
Past literature on the development of type 1 diabetes (T1D) and type 2 diabetes (T2D) has emphasized the influence of exogenous factors, including viral infections, in the development of these conditions. The coronavirus disease 2019 (COVID-19) pandemic again highlighted the complicated connection between viral infection and the development of diabetes. The complex interplay of proinflammatory, genetic, and socioeconomic factors can help explain the increased incidence of T1D and T2D during the pandemic. Proposed pathophysiological mechanisms connecting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to T1D include the expression of angiotensin enzyme 2 receptors on pancreatic islet cells, resultant proinflammatory states, and potential transient damage caused by viral entry. The intricate web of genetic factors, social determinants of health (including the rise of obesity), and the impact of proinflammatory states during SARS-CoV-2 infection on insulin resistance suggests mechanisms linking SARS-CoV-2 infection to the development of diabetes. [Pediatr Ann. 2024;53(7):e258-e263.].
Collapse
|
45
|
Espina González C, Núñez Batalla F, Mackers Iglesias P, Sumarroca Trouboul A, Cantón Bascuas M, García Lorenzo J. Dysphonia and other voice alterations associated with COVID-19: Systematic review. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2024; 75:252-259. [PMID: 38797375 DOI: 10.1016/j.otoeng.2024.02.005] [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: 10/08/2023] [Accepted: 02/21/2024] [Indexed: 05/29/2024]
Abstract
Among the symptoms presented by patients with SARS-Cov-2 infection, we can find various otorhinolaryngological alterations. Dysphonia appears in up to 79% of infected patients during the acute phase. Dysphonia can also occur as a sequelae, often underestimated, possibly due to its appearance along with other symptoms, also in patients after prolonged intubation or tracheostomy. We present a systematic review of the literature with a bibliographic search in PubMed, Cochrane and Google Scholar, with MESH terms including studies in English and Spanish. The results of the studies found and the vocal manifestations in patients during COVID-19 disease and the consequences produced are analysed. Dysphonia is an acute manifestation of COVID-19 with alterations in aerodynamic and acoustic analysis and in fibrolaryngoscopy. Post-COVID dysphonia can be a persistent symptom that is often underestimated, requiring multidisciplinary management and speech therapy intervention. Laryngeal sequelae are common in post-intubation or post-tracheostomy patients and are related to intubation time, tube number, pronation and respiratory sequelae.
Collapse
Affiliation(s)
| | - Faustino Núñez Batalla
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | | | | | | |
Collapse
|
46
|
Sideratou CM, Papaneophytou C. Persistent Vascular Complications in Long COVID: The Role of ACE2 Deactivation, Microclots, and Uniform Fibrosis. Infect Dis Rep 2024; 16:561-571. [PMID: 39051242 PMCID: PMC11270324 DOI: 10.3390/idr16040042] [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: 05/01/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2), a key regulator in vasoregulation and the renin-angiotensin system, is hypothesized to be downregulated in patients with COVID-19, leading to a cascade of cardiovascular complications. This deactivation potentially results in increased blood pressure and vessel injury, contributing to the formation and persistence of microclots in the circulation. Herein, we propose a hypothesis regarding the prolonged vascular complications observed in long COVID, focusing on the role of ACE2 deactivation and/or shedding, the persistence of microclots, and the unique pattern of fibrosis induced by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Furthermore, we propose that the distinctive, uniform fibrosis associated with COVID-19, which is challenging to detect through conventional X-ray imaging, exacerbates vascular injury and impairs oxygenation. The persistence of these microclots and the unique fibrosis pattern are suggested as key factors in the extended duration of vascular complications post-COVID-19 infection, regardless of the initial disease severity. Moreover, plasma ACE2 activity has the potential to serve as prognostic or diagnostic biomarkers for monitoring disease severity and managing long COVID symptoms. Elucidating the role of ACE2 deactivation and the consequent events is vital for understanding the long-term effects of COVID-19. The experimental verification of this hypothesis through in vitro studies, clinical longitudinal studies, and advanced imaging techniques could yield significant insights into the pathophysiological mechanisms underlying long COVID, thereby improving the management of patients, particularly those with cardiovascular complications.
Collapse
Affiliation(s)
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus;
| |
Collapse
|
47
|
Di Spigna G, Covelli B, Vargas M, Di Caprio R, Rubino V, Iacovazzo C, Napolitano F, Servillo G, Postiglione L. The Behaviour of IL-6 and Its Soluble Receptor Complex during Different Waves of the COVID-19 Pandemic. Life (Basel) 2024; 14:814. [PMID: 39063569 PMCID: PMC11278279 DOI: 10.3390/life14070814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
In late December 2019, SARS-CoV-2 was identified as the cause of a new pneumonia (COVID-19), leading to a global pandemic declared by the WHO on 11 March 2020, with significant human, economic, and social costs. Although most COVID-19 cases are asymptomatic or mild, 14% progress to severe disease, and 5% develop critical illness with complications such as interstitial pneumonia, acute respiratory distress syndrome (ARDS), and multiple organ dysfunction syndrome (MODS). SARS-CoV-2 primarily targets the respiratory system but can affect multiple organs due to the widespread presence of angiotensin-converting enzyme 2 (ACE2) receptors, which the virus uses to enter cells. This broad distribution of ACE2 receptors means that SARS-CoV-2 infection can lead to cardiovascular, gastrointestinal, renal, hepatic, central nervous system, and ocular damage. The virus triggers the innate and adaptive immune systems, resulting in a massive cytokine release, known as a "cytokine storm", which is linked to tissue damage and poor outcomes in severe lung disease. Interleukin-6 (IL-6) is particularly important in this cytokine release, with elevated levels serving as a marker of severe COVID-19. IL-6 is a multifunctional cytokine with both anti-inflammatory and pro-inflammatory properties, acting through two main pathways: classical signalling and trans-signalling. Classical signalling involves IL-6 binding to its membrane-bound receptor IL-6R and then to the gp130 protein, while trans-signalling occurs when IL-6 binds to the soluble form of IL-6R (sIL-6R) and then to membrane-bound gp130 on cells that do not express IL-6R. The soluble form of gp130 (sgp130) can inhibit IL-6 trans-signalling by binding to sIL-6R, thereby preventing it from interacting with membrane-bound gp130. Given the central role of IL-6 in COVID-19 inflammation and its association with severe disease, we aimed to analyse the behaviour of IL-6 and its soluble receptor complex during different waves of the pandemic. This analysis could help determine whether IL-6 levels can serve as prognostic markers of disease severity.
Collapse
Affiliation(s)
- Gaetano Di Spigna
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Bianca Covelli
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Maria Vargas
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.); (G.S.)
| | - Roberta Di Caprio
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Valentina Rubino
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Carmine Iacovazzo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.); (G.S.)
| | - Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| | - Giuseppe Servillo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.V.); (C.I.); (G.S.)
| | - Loredana Postiglione
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.D.S.); (B.C.); (R.D.C.); (V.R.); (F.N.)
| |
Collapse
|
48
|
Raheem MA, Rahim MA, Gul I, Reyad-Ul-Ferdous M, Zhang CY, Yu D, Pandey V, Du K, Wang R, Han S, Han Y, Qin P. COVID-19: Post infection implications in different age groups, mechanism, diagnosis, effective prevention, treatment, and recommendations. Life Sci 2024:122861. [PMID: 38925222 DOI: 10.1016/j.lfs.2024.122861] [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: 11/22/2023] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
SARS-CoV-2 is a highly contagious pathogen that predominantly caused the COVID-19 pandemic. The persistent effects of COVID-19 are defined as an inflammatory or host response to the virus that begins four weeks after initial infection and persists for an undetermined length of time. Chronic effects are more harmful than acute ones thus, this review explored the long-term effects of the virus on various human organs, including the pulmonary, cardiovascular, and neurological, reproductive, gastrointestinal, musculoskeletal, endocrine, and lymphoid systems and found that SARS-CoV-2 adversely affects these organs of older adults. Regarding diagnosis, the RT-PCR is a gold standard method of diagnosing COVID-19; however, it requires specialized equipment and personnel for performing assays and a long time for results production. Therefore, to overcome these limitations, artificial intelligence employed in imaging and microfluidics technologies is the most promising in diagnosing COVID-19. Pharmacological and non-pharmacological strategies are the most effective treatment for reducing the persistent impacts of COVID-19 by providing immunity to post-COVID-19 patients by reducing cytokine release syndrome, improving the T cell response, and increasing the circulation of activated natural killer and CD8 T cells in blood and tissues, which ultimately reduces fever, nausea, fatigue, and muscle weakness and pain. Vaccines such as inactivated viral, live attenuated viral, protein subunit, viral vectored, mRNA, DNA, or nanoparticle vaccines significantly reduce the adverse long-term virus effects in post-COVID-19 patients; however, no vaccine was reported to provide lifetime protection against COVID-19; consequently, protective measures such as physical separation, mask use, and hand cleansing are promising strategies. This review provides a comprehensive knowledge of the persistent effects of COVID-19 on people of varying ages, as well as diagnosis, treatment, vaccination, and future preventative measures against the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Muhammad Akmal Raheem
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Muhammad Ajwad Rahim
- College of Animal Science and Technology, Ahnui Agricultural University, Hefei, PR China
| | - Ijaz Gul
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Md Reyad-Ul-Ferdous
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Can Yang Zhang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Dongmei Yu
- School of Mechanical, Electrical & Information Engineering, Shandong University
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Ke Du
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA
| | - Runming Wang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Sanyang Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Yuxing Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Peiwu Qin
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China.
| |
Collapse
|
49
|
Salvio AL, Fernandes RA, Ferreira HFA, Duarte LA, Gutman EG, Raposo-Vedovi JV, Filho CHFR, da Costa Nunes Pimentel Coelho WL, Passos GF, Andraus MEC, da Costa Gonçalves JP, Cavalcanti MG, Amaro MP, Kader R, de Andrade Medronho R, Figueiredo CP, Amado-Leon LA, Alves-Leon SV. High Levels of NfL, GFAP, TAU, and UCH-L1 as Potential Predictor Biomarkers of Severity and Lethality in Acute COVID-19. Mol Neurobiol 2024; 61:3545-3558. [PMID: 37996731 PMCID: PMC11087339 DOI: 10.1007/s12035-023-03803-z] [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: 08/10/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023]
Abstract
Few studies showed that neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), total tubulin-associated unit (TAU), and ubiquitin carboxy-terminal hydrolase-L1 (UCH-L1) may be related to neurological manifestations and severity during and after SARS-CoV-2 infection. The objective of this work was to investigate the relationship among nervous system biomarkers (NfL, TAU, GFAP, and UCH-L1), biochemical parameters, and viral loads with heterogeneous outcomes in a cohort of severe COVID-19 patients admitted in Intensive Care Unit (ICU) of a university hospital. For that, 108 subjects were recruited within the first 5 days at ICU. In parallel, 16 mild COVID-19 patients were enrolled. Severe COVID-19 group was divided between "deceased" and "survivor." All subjects were positive for SARS-CoV-2 detection. NfL, total TAU, GFAP, and UCH-L1 quantification in plasma was performed using SIMOA SR-X platform. Of 108 severe patients, 36 (33.33%) presented neurological manifestation and 41 (37.96%) died. All four biomarkers - GFAP, NfL, TAU, and UCH-L1 - were significantly higher among deceased patients in comparison to survivors (p < 0.05). Analyzing biochemical biomarkers, higher Peak Serum Ferritin, D-Dimer Peak, Gamma-glutamyltransferase, and C-Reactive Protein levels were related to death (p < 0.0001). In multivariate analysis, GFAP, NfL, TAU, UCH-L1, and Peak Serum Ferritin levels were correlated to death. Regarding SARS-CoV-2 viral load, no statistical difference was observed for any group. Thus, Ferritin, NFL, GFAP, TAU, and UCH-L1 are early biomarkers of severity and lethality of SARS-COV-2 infection and may be important tools for therapeutic decision-making in the acute phase of disease.
Collapse
Affiliation(s)
- Andreza Lemos Salvio
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Renan Amphilophio Fernandes
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Helena França Alcaraz Ferreira
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Larissa Araujo Duarte
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Elisa Gouvea Gutman
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Jessica Vasques Raposo-Vedovi
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | | | | | | | - Maria Emília Cosenza Andraus
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | - João Paulo da Costa Gonçalves
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Marta Guimarães Cavalcanti
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
- Epidemiology and Evaluation Service, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | - Marisa Pimentel Amaro
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
- School of Medicine, Post-Graduate Program in Infectious and Parasitic Diseases, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | - Rafael Kader
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
- School of Medicine, Post-Graduate Program in Infectious and Parasitic Diseases, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | - Roberto de Andrade Medronho
- Epidemiology and Evaluation Service, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | | | - Luciane Almeida Amado-Leon
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil.
| | - Soniza Vieira Alves-Leon
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil.
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil.
| |
Collapse
|
50
|
Greenwald MA, Namin S, Zajdowicz J, Jones AL, Fritts L, Kuehnert MJ, Miller CJ, Ray G. Testing of tissue specimens obtained from SARS-CoV-2 nasopharyngeal swab-positive donors. Cell Tissue Bank 2024; 25:583-604. [PMID: 37995051 PMCID: PMC11143015 DOI: 10.1007/s10561-023-10119-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/20/2023] [Indexed: 11/24/2023]
Abstract
Risk for transmission of SARS-CoV-2 through allogeneic human tissue transplantation is unknown. To further evaluate the risk of virus transmission, tissues were obtained from deceased donors who had tested positive for SARS-CoV-2 RNA via nasopharyngeal swab. This study evaluated an array of human tissues recovered for transplantation, including bone, tendon, skin, fascia lata, vascular tissues, and heart valves. Tissue samples and plasma or serum samples, if available, were tested for viral RNA (vRNA) using a real time PCR system for the presence of virus RNA. All samples were tested in quadruplicate for both subgenomic (sgRNA) and genomic (gRNA) RNA encoding the SARS-CoV-2 nucleocapsid gene. Amplification of a cellular housekeeping gene served as the positive control for every sample. A total of 47 tissue samples from 17 donors were tested for SARS-CoV-2 RNA. Four donors had plasma or serum available for paired testing. SARS-CoV-2 RNA was not detected from any tissue or plasma/serum sample tested. Based on these findings, risk of transmission through the transplantation of tissue types studied from SARS-CoV-2 infected donors is likely to be low.
Collapse
Affiliation(s)
- Melissa A Greenwald
- Donor Alliance, Denver, CO, USA.
- Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | | | | | | | | | - Matthew J Kuehnert
- MTF Biologics, Edison, NJ, USA
- Hackensack Meridian School of Medicine, Hackensack, NJ, USA
| | | | | |
Collapse
|