1
|
Kaur Sardarni U, Ambikan AT, Acharya A, Johnson SD, Avedissian SN, Végvári Á, Neogi U, Byrareddy SN. Sars-Cov-2 variants mediated tissue-specific metabolomic reprogramming determines the disease pathophysiology in a hamster model. Brain Behav Immun 2024:S0889-1591(24)00674-3. [PMID: 39481495 DOI: 10.1016/j.bbi.2024.10.032] [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: 04/21/2024] [Revised: 09/28/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024] Open
Abstract
Despite significant effort, a clear understanding of host tissue-specific responses and their implications for immunopathogenicity against the severe acute respiratory syndrome coronavirus2 (SARS-CoV-2) variant infection has remained poorly defined. To shed light on the interaction between organs and specific SARS-CoV-2 variants, we sought to characterize the complex relationship among acute multisystem manifestations, dysbiosis of the gut microbiota, and the resulting implications for SARS-CoV-2 variant-specific immunopathogenesis in the Golden Syrian Hamster (GSH) model using multi-omics approaches. Our investigation revealed the presence of increased SARS-CoV-2 genomic RNA in diverse tissues of delta-infected GSH compared to the omicron variant. Multi-omics analyses uncovered distinctive metabolic responses between the delta and omicron variants, with the former demonstrating dysregulation in synaptic transmission proteins associated with neurocognitive disorders. Additionally, delta-infected GSH exhibited an altered fecal microbiota composition, marked by increased inflammation-associated taxa and reduced commensal bacteria compared to the omicron variant. These findings underscore the SARS-CoV-2-mediated tissue insult, characterized by modified host metabolites, neurological protein dysregulation, and gut dysbiosis, highlighting the compromised gut-lung-brain axis during acute infection.
Collapse
Affiliation(s)
- Urvinder Kaur Sardarni
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anoop T Ambikan
- The Systems Virology Laboratory, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Samuel D Johnson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sean N Avedissian
- Antiviral Pharmacology Laboratory, Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ujjwal Neogi
- The Systems Virology Laboratory, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Campus Flemingsberg, Stockholm, Sweden.
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
2
|
Casas Aparicio G, Fernández Plata R, Higuera Iglesias A, Martínez Briseño D, Claure-Del Granado R, Castillejos Lopez M, Vázquez Pérez J, Alvarado Vásquez N, Velázquez Cruz R, Hernández Silva G, Ruiz V, Camarena Á, Salinas Lara C, Tena Suck M, Montes de Oca Ambriz I, Ortiz Toledo O, Arvizu Serrano V, Almazan Chaparro Y, Flores-Soto E, Espíndola LMT, Aquino-Gálvez A, Ahumada Topete VH. Clinical implications of persistently increased blood urea nitrogen/serum creatinine ratio (PI-BUN/Cr) in severe COVID-19 patients. Pneumonia (Nathan) 2024; 16:20. [PMID: 39449127 PMCID: PMC11515407 DOI: 10.1186/s41479-024-00140-0] [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: 11/07/2023] [Accepted: 08/07/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Patients with COVID-19 may experience a persistent increase in the blood urea nitrogen over creatinine ratio (PI-BUN/Cr). Its elevation could reflect multiple underlying pathophysiological processes beyond prerenal injury but also warrants nuanced interpretation due to its complex interplay with various factors, underscoring the importance of investigating its effects on mortality and acute kidney injury in this population. METHODS We analized a retrospective and longitudinal cohort of patients admitted to a single center in Mexico City for patients with severe COVID-19. Between March 5, 2020 and August 25, 2021, we included patients with confirmed positive diagnosis for SARS-CoV-2, age > 18 years, disease severity was defined by clinical data of respiratory distress syndrome and a ratio of partial oxygen pressure to inspired oxygen fraction < 300 mmHg on admission. We excluded patients with End Stage Kidney Disease. Data was obtained from electronic medical records. PI-BUN/Cr was defined as an increase in the BUN/Cr ratio > 30 in more than 60% of measurements in the hospital. The outcomes included: risk factors to mortality and AKI in-hospital. RESULTS The cohort included 3,007 patients with a median age of 54.6 ± 14.5 years. 35% of patients died; 44.6% developed PI-BUN/Cr ratio and 71.4% AKI. Mortality was associated with older age > 60 years [Hazard ratio (HR)] = 1.45, 95% CI: 1.28-1.65; p < 0.001); male (HR 1.25, 95% CI 1.09-1.44; p = 0.002) and AKI (HR 3.29, 95% CI 2.42-4.46; p < 0.001); PI-BUN/CR & Non-AKI (HR = 2.82, 95% CI: 1.61-4.93; p < 0.001); Non PI-BUN/CR & AKI (HR = 5.47, 95% CI: 3.54-8.44; p < 0.001); and PI-BUN/CR & AKI (HR = 4.26, 95% CI: 2.75-6.62, p < 0.001). Only hiperuricemia was a risk factor for AKI (HR = 1.71, 95% CI: 1.30-2.25, p < 0.001). CONCLUSIONS While PI-BUN/Cr alone may not directly associate with mortality, its capacity to sub-phenotype patients according to their AKI status holds significant promise in offering valuable insights into patient prognosis and outcomes. Understanding the nuanced relationship between PI-BUN/Cr and AKI enhances our comprehension of renal function dynamics. It equips healthcare providers with a refined tool for risk stratification and personalized patient management strategies.
Collapse
Affiliation(s)
- Gustavo Casas Aparicio
- Titular de la Coordination de Nefrología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Ciudad de México, 14080, México
| | - Rosario Fernández Plata
- Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México, 14080, CP, México
| | - Anjarath Higuera Iglesias
- Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México, 14080, CP, México
| | - David Martínez Briseño
- Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México, 14080, CP, México.
| | - Rolando Claure-Del Granado
- Division de Nefrología, Cochabamba Bolivia and IIBISMED, Facultad de Medicina, Universidad Mayor de San Simón, Hospital Obrero No. 2 - CNS, Cochabamba, Bolivia
| | - Manuel Castillejos Lopez
- Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México, 14080, CP, México.
| | - Joel Vázquez Pérez
- Laboratorio de Biología Molecular de Enfermedades Emergentes y EPOC, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Ciudad de México, 14080, México
| | - Noé Alvarado Vásquez
- Departmento de Investigación en Biomedicina Molecular e Investigación Translacional, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Ciudad de México, 14080, México
| | - Rafael Velázquez Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Ciudad de México, 14610, Mexico
| | - Graciela Hernández Silva
- Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México, 14080, CP, México
| | - Victor Ruiz
- Laboratorio de Biología Molecular, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Ciudad de México, 14080, México
- SecciÓn de Estudios de Posgrado e InvestigaciÓn, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz MirÓn s/n, Col. Casco de Santo Tomas, Miguel Hidalgo, Ciudad de México, 11340, México
| | - Ángel Camarena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Ciudad de México, 14080, México
| | - Citlaltepetl Salinas Lara
- Laboratorio de Patología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velazco Suarez", Insurgentes Sur 3877, Ciudad de México, 14269, México
- Departamento de Neuropatologia, Instituto Nacional de Neurología y Neurocirugia, Manuel Velasco Suarez, Insurgentes Sur 3877, Ciudad de México, 14269, México
| | - Martha Tena Suck
- Laboratorio de Patología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velazco Suarez", Insurgentes Sur 3877, Ciudad de México, 14269, México
| | - Iñaki Montes de Oca Ambriz
- Facultad de Estudios Superiores Iztacala (Programa MEDICI), Universidad Nacional Autónoma de México, Av. de los Barrios 1, Tlalnepantla de Baz, Ciudad de México, 54090, México
| | - Oswaldo Ortiz Toledo
- Facultad de Estudios Superiores Iztacala (Programa MEDICI), Universidad Nacional Autónoma de México, Av. de los Barrios 1, Tlalnepantla de Baz, Ciudad de México, 54090, México
| | - Vianey Arvizu Serrano
- Facultad de Estudios Superiores Iztacala (Programa MEDICI), Universidad Nacional Autónoma de México, Av. de los Barrios 1, Tlalnepantla de Baz, Ciudad de México, 54090, México
| | - Yared Almazan Chaparro
- Facultad de Estudios Superiores Iztacala (Programa MEDICI), Universidad Nacional Autónoma de México, Av. de los Barrios 1, Tlalnepantla de Baz, Ciudad de México, 54090, México
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, 04510, México
| | - Luz María Torres Espíndola
- Laboratorio de Farmacología, Instituto Nacional de Pediatría, Insurgentes Sur 3700, Ciudad de México, 04530, México
| | - Arnoldo Aquino-Gálvez
- Titular de la Coordination de Nefrología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Ciudad de México, 14080, México
| | - Victor Hugo Ahumada Topete
- Unidad de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México, 14080, CP, México
| |
Collapse
|
3
|
Wesley UV, Dempsey RJ. Neuro-molecular perspectives on long COVID-19 impacted cerebrovascular diseases - a role for dipeptidyl peptidase IV. Exp Neurol 2024; 380:114890. [PMID: 39038507 DOI: 10.1016/j.expneurol.2024.114890] [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: 05/15/2024] [Revised: 07/01/2024] [Accepted: 07/14/2024] [Indexed: 07/24/2024]
Abstract
The coronavirus disease 2019 (COVID-19) has caused immense devastation globally with many outcomes that are now extending to its long-term sequel called long COVID. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects not only lungs, but also the brain and heart in association with endothelial cell dysfunction, coagulation abnormalities, and thrombosis leading to cardio-cerebrovascular health issues. Fatigue, cognitive decline, and brain fog are common neurological symptoms in persisting long COVID. Neurodegenerative processes and SARS-CoV-2 infection manifest overlapping molecular mechanisms, such as cytokine dysregulation, inflammation, protein aggregation, mitochondrial dysfunction, and oxidative stress. Identifying the key molecules in these processes is of importance for prevention and treatment of this disease. In particular, Dipeptidyl peptidase IV (DPPIV), a multifunctional peptidase has recently drawn attention as a potential co-receptor for SARS-CoV-2 infection and cellular entry. DPPIV is a known co-receptor for some other COVID viruses including MERS-Co-V. DPPIV regulates the immune responses, obesity, glucose metabolism, diabetes, and hypertension that are associated with cerebrovascular manifestations including stroke. DPPIV likely worsens persisting COVID-19 by disrupting inflammatory signaling pathways and the neurovascular system. This review highlights the neurological, cellular and molecular processes concerning long COVID, and DPPIV as a potential key factor contributing to cerebrovascular dysfunctions following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Umadevi V Wesley
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA.
| | - Robert J Dempsey
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
| |
Collapse
|
4
|
Jeon D, Kim SH, Kim J, Jeong H, Uhm C, Oh H, Cho K, Cho Y, Park IH, Oh J, Kim JJ, Hwang JY, Lee HJ, Lee HY, Seo JY, Shin JS, Seong JK, Nam KT. Discovery of a new long COVID mouse model via systemic histopathological comparison of SARS-CoV-2 intranasal and inhalation infection. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167347. [PMID: 39019092 DOI: 10.1016/j.bbadis.2024.167347] [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: 02/29/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/19/2024]
Abstract
Intranasal infection is commonly used to establish a SARS-CoV-2 mouse model due to its non-invasive procedures and a minimal effect from the operation itself. However, mice intranasally infected with SARS-CoV-2 have a high mortality rate, which limits the utility of this model for exploring therapeutic strategies and the sequelae of non-fatal COVID-19 cases. To resolve these limitations, an aerosolised viral administration method has been suggested. However, an in-depth pathological analysis comparing the two models is lacking. Here, we show that inhalation and intranasal SARS-CoV-2 (106 PFU) infection models established in K18-hACE2 mice develop unique pathological features in both the respiratory and central nervous systems, which could be directly attributed to the infection method. While the inhalation-infection model exhibited relatively milder pathological parameters, it closely mimicked the prevalent chest CT pattern observed in COVID-19 patients with focal, peripheral lesions and fibrotic scarring in the recuperating lung. We also found the evidence of direct neuron-invasion from the olfactory receptor neurons to the olfactory bulb in the intranasal model and showed the trigeminal nerve as an alternative route of transmission to the brain in inhalation infected mice. Even after viral clearance confirmed at 14 days post-infection, mild lesions were still found in the brain of inhalation-infected mice. These findings suggest that the inhalation-infection model has advantages over the intranasal-infection model in closely mimicking the pathological features of non-fatal symptoms of COVID-19, demonstrating its potential to study the sequelae and possible interventions for long COVID.
Collapse
Affiliation(s)
- Donghun Jeon
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung-Hee Kim
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Jiseon Kim
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Haengdueng Jeong
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Chanyang Uhm
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Heeju Oh
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyungrae Cho
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Yejin Cho
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - In Ho Park
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Jooyeon Oh
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jeong Jin Kim
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Yeon Hwang
- Preclinical Research Center, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hyo-Jung Lee
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea; Department of Nuclear Medicine, Seoul National University, College of Medicine, Seoul, South Korea
| | - Jun-Young Seo
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Jeon-Soo Shin
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea; Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK 21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, South Korea; BIO MAX Institute, Seoul National University, Seoul, South Korea; Interdisciplinary Program for Bioinformatics, Seoul National University, Seoul, South Korea.
| | - Ki Taek Nam
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
5
|
Singh H, Nair A, Mahajan SD. Impact of genetic variations of gene involved in regulation of metabolism, inflammation and coagulation on pathogenesis of cardiac injuries associated with COVID-19. Pathol Res Pract 2024; 263:155608. [PMID: 39447244 DOI: 10.1016/j.prp.2024.155608] [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: 07/08/2024] [Revised: 08/29/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND SARS-CoV-2 infection can result in long-term chronic cardiovascular (CV) damage after the acute phase of the illness. COVID-19 frequently causes active myocarditis, SARS-CoV-2 can directly infect and kill cardiac cells, causing severe pathology and dysfunction across the organs and cells. Till now, the pathogenesis of COVID-19-associated cardiac injuries has not been understood, but there are several factors that contribute to the progression of cardiac injuries, such as genetic, dietary, and environmental. Among them ranges of host genetic factor including metabolizing, inflammation, and coagulation related genes have a role to contribute the cardiac injuries induced by COVID-19. Hereditary DNA sequence variations contribute to the risk of illness in almost all of these diseases. Hence, we comprehended the occurrence of genetic variations of metabolizing, inflammation and coagulation-related genes in the general population, their expression in various diseases, and their impact on cardiac injuries induced by COVID-19. METHOD We utilized multiple databases, including PubMed (Medline), EMBASE, and Google Scholar, for literature searches. DESCRIPTION The genes involved in metabolism (APOE, MTHFR), coagulation (PAI-1, ACE2), and immune factors (CRP, ESR, and troponin I) may have a role in the progression of COVID-19-associated cardiac injuries. The risk factors for CVD are significantly varied between and within different regions. In healthy individuals, the ACE I allele is responsible for the predisposition to CAD, but the ACE D haplotype is responsible for susceptibility and severity, which ultimately leads to heart failure. Patients who carry the T allele of rs12329760 in the TMPRSS2 gene are at risk for developing the severe form of COVID-19. IL-6 (rs1800796/rs1800795) polymorphism is associated with an increased mortality rate and susceptibility to severe COVID-19 disease. While the putative role of IL-6 associated with chronic, inflammatory diseases like cardiac and cerebrovascular disease is well known. CONCLUSION The occurrence of genetic variations in the ACE-2, AGT, DPP-IV, TMPRSS2, FUIRN, IL-4, IL-6, IFN-γ, and CYP2D6 genes is varied among different populations. Examining the correlation between these variations and their protein levels and cardiac injuries induced by COVID-19 may provide valuable insights into the pathogenesis of cardiac injuries induced by COVID-19.
Collapse
Affiliation(s)
- HariOm Singh
- Department of Molecular Biology, National AIDS Research Institute, Pune 411026, India.
| | - Aishwarya Nair
- Department of Molecular Biology, National AIDS Research Institute, Pune 411026, India
| | - Supriya D Mahajan
- Department of Medicine, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo's Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| |
Collapse
|
6
|
Demeter F, Bihari G, Vadicsku D, Sinkovits G, Kajdácsi E, Horváth L, Réti M, Müller V, Iványi Z, Gál J, Gopcsa L, Reményi P, Szathmáry B, Lakatos B, Szlávik J, Bobek I, Prohászka ZZ, Förhécz Z, Masszi T, Vályi-Nagy I, Prohászka Z, Cervenak L. Anti-Inflammatory Cytokine Profiles in Thrombotic Thrombocytopenic Purpura-Differences Compared to COVID-19. Int J Mol Sci 2024; 25:10007. [PMID: 39337495 PMCID: PMC11432022 DOI: 10.3390/ijms251810007] [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: 08/13/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Thromboinflammation/immunothrombosis plays a role in several diseases including thrombotic thrombocytopenic purpura (TTP) and COVID-19. Unlike the extensive research that has been conducted on COVID-19 cytokine storms, the baseline and acute phase cytokine profiles of TTP are poorly characterized. Moreover, we compared the cytokine profiles of TTP and COVID-19 to identify the disease-specific/general characteristics of thromboinflammation/immunothrombosis. Plasma concentrations of 33 soluble mediators (SMs: cytokines, chemokines, soluble receptors, and growth factors) were measured by multiplex bead-based LEGENDplex™ immunoassay from 32 COVID-19 patients (32 non-vaccinated patients in three severity groups), 32 TTP patients (remission/acute phase pairs of 16 patients), and 15 control samples. Mainly, the levels of innate immunity-related SMs changed in both diseases. In TTP, ten SMs decreased in both remission and acute phases compared to the control, one decreased, and two increased only in the acute phase compared to remission, indicating mostly anti-inflammatory changes. In COVID-19, ten pro-inflammatory SMs increased, whereas one decreased with increasing severity compared to the control. In severe COVID-19, sixteen SMs exceeded acute TTP levels, with only one higher in TTP. PCA identified CXCL10, IL-1RA, and VEGF as the main discriminators among their cytokine profiles. The innate immune response is altered in both diseases. The cytokine profile of TTP suggests a distinct pathomechanism from COVID-19 and supports referring to TTP as thromboinflammatory rather than immunothrombotic, emphasizing thrombosis over inflammation as the driving force of the acute phase.
Collapse
Affiliation(s)
- Flóra Demeter
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - György Bihari
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Dorina Vadicsku
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - György Sinkovits
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Erika Kajdácsi
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
- Research Group for Immunology and Hematology, Semmelweis University—HUN-REN-SU (Office for Supported Research Groups), 1085 Budapest, Hungary
| | - Laura Horváth
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Marienn Réti
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Veronika Müller
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary
| | - Zsolt Iványi
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
| | - János Gál
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
| | - László Gopcsa
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Péter Reményi
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Beáta Szathmáry
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Botond Lakatos
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - János Szlávik
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Ilona Bobek
- Department of Anaesthesiology and Intensive Therapy, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Zita Z. Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Zsolt Förhécz
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Tamás Masszi
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - István Vályi-Nagy
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Zoltán Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
- Research Group for Immunology and Hematology, Semmelweis University—HUN-REN-SU (Office for Supported Research Groups), 1085 Budapest, Hungary
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
7
|
Valle N, Eapen MS, Pillai K, Morris R, Akhter J, Mekkawy AH, Morris DL, Valle SJ. Impact of Nebulized BromAc ® on Mucus Plug Clearance in a Mechanically Ventilated Ex Vivo Ovine Lung Model of Obstructive Respiratory Conditions. Life (Basel) 2024; 14:1111. [PMID: 39337895 PMCID: PMC11433166 DOI: 10.3390/life14091111] [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: 06/17/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Mucus plugging of the respiratory tract occurs in airway diseases, including asthma, chronic obstructive pulmonary disease, and cystic fibrosis. It can cause blockage of the airways, leading to breathlessness and lung failure. Here, we used a ventilatory setup to demonstrate the effect of BromAc® in dissolving mucus plugs in a novel ex vivo ovine obstructive lung model. Mucus simulant was filled into the trachea of freshly slaughtered ovine lungs and ventilated via an endotracheal tube (ETT) using Continuous Mandatory Ventilation. Predetermined single or repeated doses of Bromelain, Acetylcysteine (Ac), BromAc®, and saline control were administered via an Aerogen® vibrating nebulizer and ventilated for 30 or 60 min. Ventilatory recording of resistance, compliance, and tidal volume was conducted, and rheology pre- and post-treatment were measured. A significant decline in airway resistance (p < 0.0001) compared to the saline control was observed when treated with Bromelain, Ac, and BromAc®, with the latter showing a stronger mucolytic effect than single agents. The decline in resistance was also effective in shorter time points (p < 0.05) at lower doses of the drugs. Changes in compliance, peak pressure, and tidal volume were not observed after administration of the drugs. Rheology measurements revealed that BromAc®TM significantly reduced the viscosity of the mucin at the end of 30 min and 60 min time points (p < 0.001) compared to the saline control. BromAc® showed complete dissolution of the respiratory mucus simulant and improved ventilatory airflow parameters in the ex vivo ovine model.
Collapse
Affiliation(s)
- Nicole Valle
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - Mathew Suji Eapen
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - Krishna Pillai
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - Richard Morris
- Intensive Care Unit, Shoalhaven District Memorial Hospital, Nowra, NSW 2541, Australia;
| | - Javed Akhter
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - Ahmed H. Mekkawy
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - David L. Morris
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- St George and Sutherland Clinical School of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sarah J. Valle
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
- St George and Sutherland Clinical School of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Intensive Care Unit, St George Hospital, Sydney, NSW 2217, Australia
| |
Collapse
|
8
|
Milostić-Srb A, Srb N, Talapko J, Meštrović T, Žiger T, Pačarić S, Fureš R, Makarović V, Škrlec I. The Effect of COVID-19 and COVID-19 Vaccination on Assisted Human Reproduction Outcomes: A Systematic Review and Meta-Analysis. Diseases 2024; 12:201. [PMID: 39329870 PMCID: PMC11431602 DOI: 10.3390/diseases12090201] [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: 06/26/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/28/2024] Open
Abstract
The most discussed infectious disease is coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. Many research endeavors have focused on the effects of the virus on reproductive organs, as these have also been shown to carry the receptors to which the virus attaches. The results of assisted reproductive technology (ART) have been significantly affected by the pandemic, with some in vitro fertilization (IVF) centers being closed due to the risk of further spread of the disease. According to World Health Organization statistics, 17.5% of adults worldwide suffered from fertility problems in 2023; in other words, one in six people in the world have reproductive health problems. As infertility is a growing problem in the modern world and new developments in assisted reproduction are always a topic of profound interest, it is important to understand the impact of SARS-CoV-2 on reproductive health. This systematic review aimed to examine studies describing patients undergoing ART procedures with a COVID-19-positive history and to shed light on the recent evidence on the safety of COVID-19 vaccination in the ART context. A meta-analysis was conducted to confirm the results of the systematic review. The results showed a significant difference in clinical pregnancy rates between the vaccinated and unvaccinated groups and an increased miscarriage rate in those with a COVID-19-positive history. However, no significant difference in clinical pregnancy and birth rates was found in participants with a previous COVID-19 infection. The results show that further studies and research are needed, even though the spread and impact of the virus have decreased. Evidence-based information for individuals and couples undergoing infertility treatment is vital to enable informed decision-making.
Collapse
Affiliation(s)
- Andrea Milostić-Srb
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Nika Srb
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Jasminka Talapko
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Tomislav Meštrović
- University Centre Varaždin, University North, 42000 Varaždin, Croatia
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA 98195, USA
- Department for Health Metrics Sciences, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Tihomil Žiger
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Stana Pačarić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Surgery, University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Rajko Fureš
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Gynecology and Obstetrics, Zabok General Hospital and Croatian Veterans Hospital, 49210 Zabok, Croatia
| | - Vedrana Makarović
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Ivana Škrlec
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
9
|
Chang C. Immunodysregulation in immunodeficiency. Allergy Asthma Proc 2024; 45:340-346. [PMID: 39294914 DOI: 10.2500/aap.2024.45.240058] [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: 09/21/2024]
Abstract
The primary immunodeficiency diseases are often accompanied by autoimmunity, autoinflammatory, or aberrant lymphoproliferation. The paradoxical nature of this association can be explained by the multiple cells and molecules involved in immune networks that interact with each other in synergistic, redundant, antagonistic, and parallel arrangements. Because progressively more immunodeficiencies are found to have a genetic etiology, in many cases, a monogenic pathology, an understanding of why immunodeficiency is really an immune dysfunction becomes evident. Understanding the role of specific genes allows us to better understand the complete nature of the inborn error of immunity (IEI); the latter is a term generally used when a clear genetic etiology can be discerned. Autoimmune cytopenias, inflammatory bowel disease, autoimmune thyroiditis, and autoimmune liver diseases as well as lymphomas and cancers frequently accompany primary immunodeficiencies, and it is important that the practitioner be aware of this association and to expect that this is more common than not. The treatment of autoimmune or immunodysregulation in primary immunodeficiencies often involves further immunosuppression, which places the patient at even greater risk of infection. Mitigating measures to prevent such an infection should be considered as part of the treatment regimen. Treatment of immunodysregulation should be mechanism based, as much as we understand the pathways that lead to the dysfunction. Focusing on abnormalities in specific cells or molecules, e.g., cytokines, will become increasingly used to provide a targeted approach to therapy, a prelude to the success of personalized medicine in the treatment of IEIs.
Collapse
|
10
|
Phillips T, Mughrabi A, Garcia LJ, El Mouhayyar C, Hattar L, Tighiouart H, Moraco AH, Nader C, Jaber BL. Association of Body Mass Index with Multiple Organ Failure in Hospitalized Patients with COVID-19: A Multicenter Retrospective Cohort Study. J Intensive Care Med 2024; 39:768-777. [PMID: 38343031 DOI: 10.1177/08850666241232362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Purpose: This study examines whether excessive adipose tissue, as measured by the body mass index (BMI), is associated with higher systemic markers of inflammation and higher risk of severe acute organ failure among patients with coronavirus disease 2019 (COVID-19). Methods: This was a multicenter retrospective cohort study of 1370 hospitalized adults (18 years or older) with COVID-19 during the first wave of the pandemic. Patient-level variables were extracted from the electronic medical record. The primary predictor variable was the BMI at time of hospital admission, in accordance with the World Health Organization classification. Multivariable logistic regression analyses examined the association of BMI with the composite of acute respiratory distress syndrome (ARDS), as defined by the use of high-flow nasal canula, non-invasive ventilation, or mechanical ventilation, severe acute kidney injury (AKI), as defined by acute dialysis requirement, or in-hospital death. Results: After adjustment for important cofounders, the BMI stratum of > 40 kg/m2 (compared to the BMI < 25 kg/m2 reference group) was associated with higher odds for the composite of ARDS, severe AKI, or in-hospital death (adjusted odds ratio [ORadj] 1.69; 95% confidence interval [CI]1.03, 2.78). As a continuous variable, BMI (per 5-kg/m2 increase) remained independently associated with the composite outcome (ORadj 1.13; 95% CI 1.03, 1.23); patients in higher BMI categories exhibited significantly higher peak levels of C-reactive protein (CRP), a systemic marker of inflammation (P = .01). In a sub-cohort of 889 patients, the association of BMI with the composite outcome was no longer significant after adjustment for the peak level of CRP. Conclusions: Among hospitalized patients with COVID-19, a higher BMI is associated with higher risk of severe organ failure or in-hospital death, which dissipates after adjustment for CRP level. This supports the hypothesis that inflammation is a downstream mediator of adipose tissue on acute organ dysfunction.
Collapse
Affiliation(s)
- Timothy Phillips
- Department of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Abdallah Mughrabi
- Department of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Levindo J Garcia
- Department of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Christopher El Mouhayyar
- Department of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Laith Hattar
- Department of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Hocine Tighiouart
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
- Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA, USA
| | - Andrew H Moraco
- Department of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
| | - Claudia Nader
- Department of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
- Division of Infectious Disease, St. Elizabeth's Medical Center, Boston, MA, USA
| | - Bertrand L Jaber
- Department of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
- Division of Nephrology, St. Elizabeth's Medical Center, Boston, MA, USA
| |
Collapse
|
11
|
Ursescu C, Teodoru G, Bucurica S, Nica RI, Lazăr ȘD, Popescu MN, Ciobanu I, Berteanu M. Using the ClinFIT COVID-19 Instrument to Assess the Functional Impairments Specific to Post-COVID-19 Patients in Romania. Diagnostics (Basel) 2024; 14:1540. [PMID: 39061679 PMCID: PMC11275825 DOI: 10.3390/diagnostics14141540] [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: 06/03/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION The COVID-19 pandemic has led to approximately 3.5 million cases in Romania, causing systemic inflammation and over 200 symptoms affecting various body systems. This complexity has challenged rehabilitation systems, necessitating personalized plans tailored to each patient's illness stage and impairment level. The ISPRM-developed ClinFIT COVID-19 instrument, aligned with the ICF categories, assists in assessing patients during acute, post-acute, and long-term phases. OBJECTIVE This study aimed to evaluate and assess functional impairments in post-COVID-19 patients in Romania, with a secondary goal of generating rehabilitation directions. METHODS Data were collected from patients at two Bucharest medical centers, including those with persistent symptoms post-acute phase. Participants were assessed using the adapted ClinFIT COVID-19 instrument, and descriptive statistics were applied. CONCLUSIONS Findings revealed diverse functional impairments in physical, psychological, and social domains among post-COVID-19 patients, with severe impairments more common in those with long-term COVID-19. Complete impairment in complex movement and paid work was noted, affecting one-third of salaried employees and forcing some to retire. In the acute phase, the most frequent functional impairments were sleep, attention, pain sensation, and exercise tolerance functions. In contrast, the most severely affected functions were exercise tolerance and mobility joint functions. Age did not positively correlate with any of the analyzed functions. In the post-acute phase, sleep, energy, and drive functions remained the most frequently affected functions, while the most severely affected was, by far, the moving around function. In the post-acute period, respiratory and respiratory muscle functions strongly correlated with all tasks related to physical activity. In the long COVID-19 phase, remunerative employment was the most severely affected function, while attention functions remained the most frequently affected, similar to the acute phase. The ClinFIT COVID-19 instrument effectively captured these impairments, underscoring the need for comprehensive rehabilitation strategies.
Collapse
Affiliation(s)
- Clara Ursescu
- Department of Rehabilitation Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Rehabilitation Medicine, University Emergency Central Military Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Gigi Teodoru
- Department of Rehabilitation Medicine, University Emergency Central Military Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Sandica Bucurica
- Department of Internal Medicine and Gastroenterology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Gastroenterology, University Emergency Central Military Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Remus Iulian Nica
- Discipline of General Surgery, Faculty of Midwifery and Nursing, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of General Surgery, University Emergency Central Military Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Ștefan Dragoș Lazăr
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Marius Nicolae Popescu
- Department of Physical and Rehabilitation Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.N.P.); (I.C.); (M.B.)
| | - Ileana Ciobanu
- Department of Physical and Rehabilitation Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.N.P.); (I.C.); (M.B.)
| | - Mihai Berteanu
- Department of Physical and Rehabilitation Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.N.P.); (I.C.); (M.B.)
| |
Collapse
|
12
|
Gupta P, Gupta A, Bansal S, Balakrishnan I. Cardiac troponin in hospitalized COVID-19 patients: Incidence, predictors, and outcomes. Ann Clin Biochem 2024; 61:255-264. [PMID: 37944990 DOI: 10.1177/00045632231216599] [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: 11/12/2023]
Abstract
BACKGROUND The incidence, predictors, and association of cardiac troponin with mortality in hospitalized COVID-19 were not adequately studied in the past and were also not reported from an Indian hospital. METHODS In this retrospective cohort study, the cardiac troponin of 240 hospitalized COVID-19 patients was measured. The incidence, predictors, and association of elevated cardiac troponin with in-hospital mortality were determined among hospitalized COVID-19 patients. RESULTS The cardiac troponin was elevated in 12.9% (31/240) of the patients. The troponin was elevated in the patients in the older age group (64 years vs. 55 years, p = .002), severe COVID-19 illness (SpO2 < 90%) (93.5% vs. 60.8%, p < .001), low arterial oxygen saturation (SpO2) (80% vs. 88%, p = .001), and low PaO2/FiO2 ratio (p < .0001). The patients with elevated cardiac troponin had elevated total leukocyte counts (TLC) (p = .001), liver enzyme (p = .025), serum creatinine (p = .011), N-terminal-Pro Brain natriuretic peptide (p < .0001), and d-dimer (p < .0001). The majority of the patients with elevated cardiac troponin were admitted to the intensive care unit (90.3% vs. 51.2%; p < .0001), were on a ventilator (61.3% vs. 21.5%; p < .0001), and had higher mortality (64.5% vs. 19.6%; p < .0001). The Kaplan-Meir survival analysis showed that the patients with elevated troponin had worse survival (p log-rank<.0001). Age, NT-ProBNP, d-dimer, and ventilator were the predictors of elevated troponin in multivariate logistic regression analysis. The Cox-regression analysis showed a significant association between elevated cardiac troponin and in-hospital mortality (adjusted hazard ratio 2.13; 95% confidence interval [CI] 1.145-3.97; p = .017). Two-thirds (65%) of patients with elevated cardiac troponin died during their hospital stay. CONCLUSIONS COVID-19 patients with elevated cardiac troponin had severe COVID illness, were more commonly admitted to an intensive care unit, were on a ventilator, and had high in-hospital mortality.
Collapse
Affiliation(s)
- Praveen Gupta
- Department of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Anunay Gupta
- Department of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Sandeep Bansal
- Department of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Ira Balakrishnan
- Department of Anesthesia and Critical Care, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| |
Collapse
|
13
|
Srivastava A, Nalroad Sundararaj S, Bhatia J, Singh Arya D. Understanding long COVID myocarditis: A comprehensive review. Cytokine 2024; 178:156584. [PMID: 38508059 DOI: 10.1016/j.cyto.2024.156584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/21/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
Infectious diseases are a cause of major concern in this twenty-first century. There have been reports of various outbreaks like severe acute respiratory syndrome (SARS) in 2003, swine flu in 2009, Zika virus disease in 2015, and Middle East Respiratory Syndrome (MERS) in 2012, since the start of this millennium. In addition to these outbreaks, the latest infectious disease to result in an outbreak is the SARS-CoV-2 infection. A viral infection recognized as a respiratory illness at the time of emergence, SARS-CoV-2 has wreaked havoc worldwide because of its long-lasting implications like heart failure, sepsis, organ failure, etc., and its significant impact on the global economy. Besides the acute illness, it also leads to symptoms months later which is called long COVID or post-COVID-19 condition. Due to its ever-increasing prevalence, it has been a significant challenge to treat the affected individuals and manage the complications as well. Myocarditis, a long-term complication of coronavirus disease 2019 (COVID-19) is an inflammatory condition involving the myocardium of the heart, which could even be fatal in the long term in cases of progression to ventricular dysfunction and heart failure. Thus, it is imperative to diagnose early and treat this condition in the affected individuals. At present, there are numerous studies which are in progress, investigating patients with COVID-19-related myocarditis and the treatment strategies. This review focuses primarily on myocarditis, a life-threatening complication of COVID-19 illness, and endeavors to elucidate the pathogenesis, biomarkers, and management of long COVID myocarditis along with pipeline drugs in detail.
Collapse
Affiliation(s)
- Arti Srivastava
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Jagriti Bhatia
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
14
|
Al-Hassinah S, Al-Daihan S, Alahmadi M, Alghamdi S, Almulhim R, Obeid D, Arabi Y, Alswaji A, Aldriwesh M, Alghoribi M. Interplay of Demographic Influences, Clinical Manifestations, and Longitudinal Profile of Laboratory Parameters in the Progression of SARS-CoV-2 Infection: Insights from the Saudi Population. Microorganisms 2024; 12:1022. [PMID: 38792852 PMCID: PMC11124088 DOI: 10.3390/microorganisms12051022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Understanding the factors driving SARS-CoV-2 infection progression and severity is complex due to the dynamic nature of human physiology. Therefore, we aimed to explore the severity risk indicators of SARS-CoV-2 through demographic data, clinical manifestations, and the profile of laboratory parameters. The study included 175 patients either hospitalized at King Abdulaziz Medical City-Riyadh or placed in quarantine at designated hotels in Riyadh, Saudi Arabia, from June 2020 to April 2021. Hospitalized patients were followed up through the first week of admission. Demographic data, clinical presentations, and laboratory results were retrieved from electronic patient records. Our results revealed that older age (OR: 1.1, CI: [1.1-1.12]; p < 0.0001), male gender (OR: 2.26, CI: [1.0-5.1]; p = 0.047), and blood urea nitrogen level (OR: 2.56, CI: [1.07-6.12]; p = 0.034) were potential predictors of severity level. In conclusion, the study showed that apart from laboratory parameters, age and gender could potentially predict the severity of SARS-CoV-2 infection in the early stages. To our knowledge, this study is the first in Saudi Arabia to explore the longitudinal profile of laboratory parameters among risk factors, shedding light on SARS-CoV-2 infection progression parameters.
Collapse
Affiliation(s)
- Sarah Al-Hassinah
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Sooad Al-Daihan
- Biochemistry Department, College of Science, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Mashael Alahmadi
- Research Office, Saudi National Institute of Health (SNIH), Riyadh 12382, Saudi Arabia;
| | - Sara Alghamdi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
| | - Rawabi Almulhim
- Infection Prevention and Control Department, King Abdulaziz Medical City, Riyadh 14611, Saudi Arabia;
| | - Dalia Obeid
- King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia;
| | - Yaseen Arabi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Intensive Care Department, King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Abdulrahman Alswaji
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
| | - Marwh Aldriwesh
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Majed Alghoribi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, Riyadh 11426, Saudi Arabia; (S.A.-H.); (S.A.); (Y.A.); (A.A.); (M.A.)
- Department of Basic Science, College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| |
Collapse
|
15
|
Lourenço AA, Amaral PHR, Paim AAO, Marques GF, Gomes-de-Pontes L, da Mata CPSM, da Fonseca FG, Pérez JCG, Coelho-dos-Reis JGA. Algorithms for predicting COVID outcome using ready-to-use laboratorial and clinical data. Front Public Health 2024; 12:1347334. [PMID: 38807995 PMCID: PMC11130428 DOI: 10.3389/fpubh.2024.1347334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
The pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging crisis affecting the public health system. The clinical features of COVID-19 can range from an asymptomatic state to acute respiratory syndrome and multiple organ dysfunction. Although some hematological and biochemical parameters are altered during moderate and severe COVID-19, there is still a lack of tools to combine these parameters to predict the clinical outcome of a patient with COVID-19. Thus, this study aimed at employing hematological and biochemical parameters of patients diagnosed with COVID-19 in order to build machine learning algorithms for predicting COVID mortality or survival. Patients included in the study had a diagnosis of SARS-CoV-2 infection confirmed by RT-PCR and biochemical and hematological measurements were performed in three different time points upon hospital admission. Among the parameters evaluated, the ones that stand out the most are the important features of the T1 time point (urea, lymphocytes, glucose, basophils and age), which could be possible biomarkers for the severity of COVID-19 patients. This study shows that urea is the parameter that best classifies patient severity and rises over time, making it a crucial analyte to be used in machine learning algorithms to predict patient outcome. In this study optimal and medically interpretable machine learning algorithms for outcome prediction are presented for each time point. It was found that urea is the most paramount variable for outcome prediction over all three time points. However, the order of importance of other variables changes for each time point, demonstrating the importance of a dynamic approach for an effective patient's outcome prediction. All in all, the use of machine learning algorithms can be a defining tool for laboratory monitoring and clinical outcome prediction, which may bring benefits to public health in future pandemics with newly emerging and reemerging SARS-CoV-2 variants of concern.
Collapse
Affiliation(s)
- Alice Aparecida Lourenço
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Adriana Alves Oliveira Paim
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovane Ferreira Marques
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leticia Gomes-de-Pontes
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- CT Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juan Carlos González Pérez
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jordana Grazziela Alves Coelho-dos-Reis
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
16
|
Sartini S, Ferrari L, Cutuli O, Castellani L, Cristina ML, Arboscello E, Sartini M. The Role of POCUS to Face COVID-19: A Narrative Review. J Clin Med 2024; 13:2756. [PMID: 38792298 PMCID: PMC11121862 DOI: 10.3390/jcm13102756] [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: 04/02/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
COVID-19 has been a challenging outbreak to face, with millions of deaths among the globe. Acute respiratory failure due to interstitial pneumonia was the leading cause of death other than prothrombotic activation and complications. Lung ultrasound (LUS) and point-of-care ultrasound (POCUS) are widely used not only to triage, to identify, and to monitor lungs involvement but also to assess hemodynamic status and thrombotic and hemorrhagic complications, mainly in critically ill patients. POCUS has gained growing consideration due to its bedside utilization, reliability, and reproducibility even in emergency settings especially in unstable patients. In this narrative review, we aim to describe LUS and POCUS utilization in COVID-19 infection based on the literature found on this topic. We reported the LUS patterns of COVID-19 pulmonary infection, the diagnostic accuracy with respect to CT lung scan, its prognostic value, the variety of scores and protocols proposed, and the utilization of POCUS to investigate the extra-lung complications.
Collapse
Affiliation(s)
- Stefano Sartini
- Emergency Medicine Department, IRCCS Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; (O.C.); (L.C.); (E.A.)
| | - Lorenzo Ferrari
- Emergency Medicine Post-Graduate School, University of Genoa, Via Balbi 5, 16126 Genoa, Italy;
| | - Ombretta Cutuli
- Emergency Medicine Department, IRCCS Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; (O.C.); (L.C.); (E.A.)
| | - Luca Castellani
- Emergency Medicine Department, IRCCS Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; (O.C.); (L.C.); (E.A.)
| | - Maria Luisa Cristina
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy;
- Hospital Hygiene, E.O. Ospedali Galliera, Via Alessandro Volta 8, 16128 Genoa, Italy
| | - Eleonora Arboscello
- Emergency Medicine Department, IRCCS Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; (O.C.); (L.C.); (E.A.)
| | - Marina Sartini
- Department of Health Sciences, University of Genoa, 16132 Genoa, Italy;
- Hospital Hygiene, E.O. Ospedali Galliera, Via Alessandro Volta 8, 16128 Genoa, Italy
| |
Collapse
|
17
|
Nakai T, Son D. Acute Mesenteric Ischemia Mimicking Severe COVID-19 Pneumonia: A Case Report on Diagnostic Challenges and Management. Cureus 2024; 16:e60466. [PMID: 38882959 PMCID: PMC11180220 DOI: 10.7759/cureus.60466] [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] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
This article demonstrates the need for abdominal imaging in COVID-19 patients with systemic symptoms in the differential diagnosis of acute mesenteric ischemia and critical COVID-19 pneumonia. We detail the case of a 91-year-old man, initially diagnosed with severe COVID-19 pneumonia, who was later found to have acute mesenteric ischemia through abdominal CT imaging, despite lacking typical abdominal symptoms. Abdominal CT revealed intramural and portal emphysema, leading to a diagnosis of acute mesenteric ischemia. Given the patient's advanced age and poor condition, supportive care was chosen, with the patient passing away 12 hours post-admission. This case highlights the critical need for comprehensive evaluation, including abdominal imaging, in COVID-19 patients with systemic symptoms to identify other serious conditions like acute mesenteric ischemia, especially in the absence of specific abdominal pain. Early detection is vital for appropriate management and improved patient outcomes.
Collapse
Affiliation(s)
- Tsubasa Nakai
- Department of Community-Based Family Medicine, Faculty of Medicine, Tottori University, Yonago, JPN
- Department of General Medicine, Hino Hospital Association Hino Hospital, Hino-gun, JPN
| | - Daisuke Son
- Department of Community-Based Family Medicine, Faculty of Medicine, Tottori University, Yonago, JPN
| |
Collapse
|
18
|
Tsiatsiou P, Kouirouxis K, Tsaireli V, Lanta A, Kassomenaki A, Papaioannou M, Protonotariou E, Skoura L. Angiopoietins as Predictor Indexes in COVID-19 Patients in Delta and Omicron Waves. Curr Issues Mol Biol 2024; 46:3975-3989. [PMID: 38785513 PMCID: PMC11120536 DOI: 10.3390/cimb46050245] [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: 02/18/2024] [Revised: 04/07/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
This study aimed to explore the correlation between Angiopoietin-1 (Ang-1) and Angiopoietin-2 (Ang-2) concentrations and the Angiopoietin-2/Angiopoietin-1 ratio (Ang-2/Ang-1) with clinical outcomes, potentially serving as disease severity and survival biomarkers. A study at AHEPA University Hospital involved 90 Coronavirus Disease 2019 (COVID-19) adult patients, 30 hospitalized intensive care units (ICU), 30 inward units (non-ICU), and 30 asymptomatic non-hospitalized individuals as controls. Estimated endothelial dysfunction markers related to angiogenesis were measured. There was a statistically significant difference only between outpatient and hospitalized patients (non-ICU-ICU groups) for the Ang-1 and Ang-2 indices. The Ang-2/Ang-1 ratio has differed significantly among the individual patient groups. An ROC analysis was conducted to find an optimal threshold for distinguishing between (outpatients-non-ICU) and (non-ICU-ICU) groups. It was based on Youden's index of 0.1122 and 0.3825, respectively. The Ang-1, Ang-2 levels, and Ang-2/Ang-1 ratio were analyzed as severity indicators in COVID-19 patients. The Ang-2/Ang-1 ratio demonstrated better prognostic and diagnostic utility than individual biomarker levels. Monitoring the Ang-2/Ang-1 ratio can identify COVID-19 patients at risk and assist clinicians in tailoring treatment strategies to improve outcomes.
Collapse
Affiliation(s)
- Panagiota Tsiatsiou
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Kyriakos Kouirouxis
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Vasiliki Tsaireli
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Antonia Lanta
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Angeliki Kassomenaki
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Maria Papaioannou
- Division of Hematology, First Department of Internal Medicine, AHEPA General Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Efthymia Protonotariou
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| | - Lemonia Skoura
- Department of Microbiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.K.); (V.T.); (A.L.); (A.K.); (E.P.); (L.S.)
| |
Collapse
|
19
|
Gong P, Zhang R, Xiao K, Shu H, Li X, Fan H, Sun X. DNA G-Quadruplex in NRP1 Promoter Facilitates SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:4422. [PMID: 38674009 PMCID: PMC11050221 DOI: 10.3390/ijms25084422] [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: 03/24/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection continues to raise concerns worldwide. Numerous host factors involved in SARS-CoV-2 infection have been identified, but the regulatory mechanisms of these host factor remain unclear. Here, we report the role of G-quadruplexes (G4s) located in the host factor promoter region in SARS-CoV-2 infection. Using bioinformatics, biochemical, and biological assays, we provide evidence for the presence of G4 structures in the promoter regions of SARS-CoV-2 host factors NRP1. Specifically, we focus on two representative G4s in the NRP1 promoter and highlight its importance in SARS-CoV-2 pathogenesis. The presence of the G4 structure greatly increases NRP1 expression, facilitating SARS-CoV-2 entry into cells. Utilizing published single-cell RNA sequencing data obtained from simulated SARS-CoV-2 infection in human bronchial epithelial cells (HBECs), we found that ciliated cells with high levels of NRP1 are prominently targeted by the virus during infection. Furthermore, our study identifies E2F1 act as a transcription factor that binds to G4s. These findings uncover a previously unknown mechanism underlying SARS-CoV-2 infection and suggest that targeting G4 structures could be a potential strategy for COVID-19 prevention and treatment.
Collapse
Affiliation(s)
- Pihai Gong
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Rongxin Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Ke Xiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Huiling Shu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Xinxiu Li
- Department of Medical Genetics and Developmental Biology, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China;
| | - Hong Fan
- Department of Medical Genetics and Developmental Biology, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China;
| | - Xiao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
20
|
Alsayari BM, Alshehri SM, Almulhim AY, Alzakry LM, Alzuraiq AA, Binshalhoub FH, Banjer HM, Alkhediwi LMA, Rasdwi KM, Khan AS. COVID-19 and Its Impact on Back Pain in the Eastern Province of Saudi Arabia. Cureus 2024; 16:e57475. [PMID: 38699131 PMCID: PMC11065479 DOI: 10.7759/cureus.57475] [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] [Accepted: 03/31/2024] [Indexed: 05/05/2024] Open
Abstract
Background Low back pain (LBP) is a common musculoskeletal condition that affects individuals worldwide, causing difficulties in daily tasks and social interactions. It can be categorized based on chronicity, with acute, subacute, and chronic forms. The causes of backache vary among patients and can include inflammatory conditions, radiculopathy, pregnancy, trauma, osteoporosis, nerve root compression, cancer, plexopathy, infection, and other spinal diseases. Aim The aim is to investigate the association between COVID-19 infection and LBP between all Saudi adults and foreign adults who had positive COVID-19 tests in the eastern region of Saudi Arabia. Methods A cross-sectional study was conducted in the Eastern Province of Saudi Arabia over the period from March 2023 to August 2023. Participants were selected by using a convenience sampling method, a sample (n=500) of individuals. The structured questionnaire was used to gather information on sociodemographic variables and COVID-related features. All the statistical calculations were performed using the SPSS software (by IBM) version 29.0.0. Results 482 participants completed the questionnaire. Out of 482 participants, the majority were females with a number of 372 (77.2%) aged between 20 and 29 years (38.4%). Out of the remaining participants, 110 (22.8%) were males. Most of the participants with a number of 301 (62.4%) were from the Hasa province. This was followed by Qatif (79, 16.4%), Dammam (56, 11.6%), Jubail (25, 5.2%), and others (21, 4.4%). The study revealed that 10.1% of participants reported experiencing back pain. The duration of backaches varied among respondents, with 122 (25.3%) experiencing them from a day to a week, 28 (5.8%) enduring them for six weeks, and 65 (13.5%) reporting a duration of six to 12 weeks. The majority, comprising 267 (55.4%) respondents, were uncertain about the period of their backaches. The prevalence of COVID-19 infection among the participants was 357 (74.1%), and 477 (99.0%) had been vaccinated against COVID-19. Approximately 44.4% of the participants experienced back pain, and out of those, 28.2% reported having pain during quarantine. Among the individuals with back pain, 24.7% attributed it to COVID-19. Conclusion This study highlights the significant correlation between back pain and COVID-19, even after the resolution of other symptoms. It underscores the importance of further research into the long-term effects and mechanisms of this association. The findings emphasize the need for healthcare professionals to consider back pain as a potential aspect of the post-COVID-19 symptom profile, ensuring comprehensive care for affected individuals.
Collapse
Affiliation(s)
| | | | | | - Leena M Alzakry
- Medicine, Imam Muhammad Ibn Saud Islamic University, Riyadh, SAU
| | | | - Fahad H Binshalhoub
- Medicine and Surgery, Imam Muhammad Ibn Saud Islamic University, Riyadh, SAU
| | - Hanin M Banjer
- College of Medicine, King Abdulaziz University, Jeddah, SAU
| | | | | | | |
Collapse
|
21
|
Ferous S, Siafakas N, Boufidou F, Patrinos GP, Tsakris A, Anastassopoulou C. Investigating ABO Blood Groups and Secretor Status in Relation to SARS-CoV-2 Infection and COVID-19 Severity. J Pers Med 2024; 14:346. [PMID: 38672973 PMCID: PMC11051264 DOI: 10.3390/jpm14040346] [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: 02/20/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
The ABO blood groups, Lewis antigens, and secretor systems are important components of transfusion medicine. These interconnected systems have been also shown to be associated with differing susceptibility to bacterial and viral infections, likely as the result of selection over the course of evolution and the constant tug of war between humans and infectious microbes. This comprehensive narrative review aimed to explore the literature and to present the current state of knowledge on reported associations of the ABO, Lewis, and secretor blood groups with SARS-CoV-2 infection and COVID-19 severity. Our main finding was that the A blood group may be associated with increased susceptibility to SARS-CoV-2 infection, and possibly also with increased disease severity and overall mortality. The proposed pathophysiological pathways explaining this potential association include antibody-mediated mechanisms and increased thrombotic risk amongst blood group A individuals, in addition to altered inflammatory cytokine expression profiles. Preliminary evidence does not support the association between ABO blood groups and COVID-19 vaccine response, or the risk of developing long COVID. Even though the emergency state of the pandemic is over, further research is needed especially in this area since tens of millions of people worldwide suffer from lingering COVID-19 symptoms.
Collapse
Affiliation(s)
- Stefanos Ferous
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (S.F.); (A.T.)
| | - Nikolaos Siafakas
- Department of Clinical Microbiology, Attikon General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Fotini Boufidou
- Neurochemistry and Biological Markers Unit, 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - George P. Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece;
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (S.F.); (A.T.)
| | - Cleo Anastassopoulou
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (S.F.); (A.T.)
| |
Collapse
|
22
|
Giovanetti M, Pannella G, Altomare A, Rocchi G, Guarino M, Ciccozzi M, Riva E, Gherardi G. Exploring the Interplay between COVID-19 and Gut Health: The Potential Role of Prebiotics and Probiotics in Immune Support. Viruses 2024; 16:370. [PMID: 38543736 PMCID: PMC10975078 DOI: 10.3390/v16030370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 05/23/2024] Open
Abstract
The COVID-19 pandemic has profoundly impacted global health, leading to extensive research focused on developing strategies to enhance outbreak response and mitigate the disease's severity. In the aftermath of the pandemic, attention has shifted towards understanding and addressing long-term health implications, particularly in individuals experiencing persistent symptoms, known as long COVID. Research into potential interventions to alleviate long COVID symptoms has intensified, with a focus on strategies to support immune function and mitigate inflammation. One area of interest is the gut microbiota, which plays a crucial role in regulating immune responses and maintaining overall health. Prebiotics and probiotics, known for their ability to modulate the gut microbiota, have emerged as potential therapeutic agents in bolstering immune function and reducing inflammation. This review delves into the intricate relationship between long COVID, the gut microbiota, and immune function, with a specific focus on the role of prebiotics and probiotics. We examine the immune response to long COVID, emphasizing the importance of inflammation and immune regulation in the persistence of symptoms. The potential of probiotics in modulating immune responses, including their mechanisms in combating viral infections such as COVID-19, is discussed in detail. Clinical evidence supporting the use of probiotics in managing long COVID symptoms is summarized, highlighting their role as adjunctive therapy in addressing various aspects of SARS-CoV-2 infection and its aftermath.
Collapse
Affiliation(s)
- Marta Giovanetti
- Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Roma, Italy; (G.P.); (A.A.)
- Climate Amplified Diseases and Epidemics (CLIMADE), Brasilia 70070-130, Brazil
- Instituto Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
| | - Gianfranco Pannella
- Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Roma, Italy; (G.P.); (A.A.)
- Department of Agricultural, Enviromental and Food Science, University of Molise, 86100 Campobasso, Italy
| | - Annamaria Altomare
- Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Roma, Italy; (G.P.); (A.A.)
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (G.R.); (M.G.)
| | - Giulia Rocchi
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (G.R.); (M.G.)
| | - Michele Guarino
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (G.R.); (M.G.)
- Operative Research Unit of Gastroenterology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128 Roma, Italy;
| | - Elisabetta Riva
- Unit of Virology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
- Applied Bacteriological Sciences Unit, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Giovanni Gherardi
- Applied Bacteriological Sciences Unit, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| |
Collapse
|
23
|
Ruz-Maldonado I, Gonzalez JT, Zhang H, Sun J, Bort A, Kabir I, Kibbey RG, Suárez Y, Greif DM, Fernández-Hernando C. Heterogeneity of hepatocyte dynamics restores liver architecture after chemical, physical or viral damage. Nat Commun 2024; 15:1247. [PMID: 38341404 PMCID: PMC10858916 DOI: 10.1038/s41467-024-45439-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Midlobular hepatocytes are proposed to be the most plastic hepatic cell, providing a reservoir for hepatocyte proliferation during homeostasis and regeneration. However, other mechanisms beyond hyperplasia have been little explored and the contribution of other hepatocyte subpopulations to regeneration has been controversial. Thus, re-examining hepatocyte dynamics during regeneration is critical for cell therapy and treatment of liver diseases. Using a mouse model of hepatocyte- and non-hepatocyte- multicolor lineage tracing, we demonstrate that midlobular hepatocytes also undergo hypertrophy in response to chemical, physical, and viral insults. Our study shows that this subpopulation also combats liver impairment after infection with coronavirus. Furthermore, we demonstrate that pericentral hepatocytes also expand in number and size during the repair process and Galectin-9-CD44 pathway may be critical for driving these processes. Notably, we also identified that transdifferentiation and cell fusion during regeneration after severe injury contribute to recover hepatic function.
Collapse
Affiliation(s)
- Inmaculada Ruz-Maldonado
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, 06520, USA
- Departments of Internal Medicine (Endocrinology) and Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - John T Gonzalez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Hanming Zhang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jonathan Sun
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Alicia Bort
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Inamul Kabir
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06511, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Richard G Kibbey
- Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, 06520, USA
- Departments of Internal Medicine (Endocrinology) and Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
- Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Daniel M Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06511, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Yale Center of Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, 06520, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
24
|
Pathak R, Eliscovich C, Mena I, Cupic A, Rutkowska M, Chandran K, Jangra RK, García-Sastre A, Singer RH, Kalpana GV. Visualization of Early RNA Replication Kinetics of SARS-CoV-2 by Using Single Molecule RNA-FISH Combined with Immunofluorescence. Viruses 2024; 16:262. [PMID: 38400039 PMCID: PMC10893374 DOI: 10.3390/v16020262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
SARS-CoV-2 infection remains a global burden. Despite intensive research, the mechanism and dynamics of early viral replication are not completely understood, such as the kinetics of the formation of genomic RNA (gRNA), sub-genomic RNA (sgRNA), and replication centers/organelles (ROs). We employed single-molecule RNA-fluorescence in situ hybridization (smRNA-FISH) to simultaneously detect viral gRNA and sgRNA and immunofluorescence to detect nsp3 protein, a marker for the formation of RO, and carried out a time-course analysis. We found that single molecules of gRNA are visible within the cytoplasm at 30 min post infection (p.i.). Starting from 2 h p.i., most of the viral RNA existed in clusters/speckles, some of which were surrounded by single molecules of sgRNA. These speckles associated with nsp3 protein starting at 3 h p.i., indicating that these were precursors to ROs. Furthermore, RNA replication was asynchronous, as cells with RNA at all stages of replication were found at any given time point. Our probes detected the SARS-CoV-2 variants of concern, and also suggested that the BA.1 strain exhibited a slower rate of replication kinetics than the WA1 strain. Our results provide insights into the kinetics of SARS-CoV-2 early post-entry events, which will facilitate identification of new therapeutic targets for early-stage replication to combat COVID-19.
Collapse
Affiliation(s)
- Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.C.); (R.K.J.)
| | - Carolina Eliscovich
- Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY 10461, USA;
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (I.M.); (A.C.); (M.R.); (A.G.-S.)
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anastasija Cupic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (I.M.); (A.C.); (M.R.); (A.G.-S.)
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Magdalena Rutkowska
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (I.M.); (A.C.); (M.R.); (A.G.-S.)
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.C.); (R.K.J.)
| | - Rohit K. Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.C.); (R.K.J.)
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (I.M.); (A.C.); (M.R.); (A.G.-S.)
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert H. Singer
- Departments of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
- Departments of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ganjam V. Kalpana
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.C.); (R.K.J.)
| |
Collapse
|
25
|
Rosas-Murrieta NH, Rodríguez-Enríquez A, Herrera-Camacho I, Millán-Pérez-Peña L, Santos-López G, Rivera-Benítez JF. Comparative Review of the State of the Art in Research on the Porcine Epidemic Diarrhea Virus and SARS-CoV-2, Scope of Knowledge between Coronaviruses. Viruses 2024; 16:238. [PMID: 38400014 PMCID: PMC10892376 DOI: 10.3390/v16020238] [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: 12/13/2023] [Revised: 01/17/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
This review presents comparative information corresponding to the progress in knowledge of some aspects of infection by the porcine epidemic diarrhea virus (PEDV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) coronaviruses. PEDV is an alphacoronavirus of great economic importance due to the million-dollar losses it generates in the pig industry. PEDV has many similarities to the SARS-CoV-2 betacoronavirus that causes COVID-19 disease. This review presents possible scenarios for SARS-CoV-2 based on the collected literature on PEDV and the tools or strategies currently developed for SARS-CoV-2 that would be useful in PEDV research. The speed of the study of SARS-CoV-2 and the generation of strategies to control the pandemic was possible due to the knowledge derived from infections caused by other human coronaviruses such as severe acute respiratory syndrome (SARS) and middle east respiratory syndrome (MERS). Therefore, from the information obtained from several coronaviruses, the current and future behavior of SARS-CoV-2 could be inferred and, with the large amount of information on the virus that causes COVID-19, the study of PEDV could be improved and probably that of new emerging and re-emerging coronaviruses.
Collapse
Affiliation(s)
- Nora H. Rosas-Murrieta
- Centro de Química, Laboratorio de Bioquímica y Biología Molecular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (A.R.-E.); (I.H.-C.); (L.M.-P.-P.)
| | - Alan Rodríguez-Enríquez
- Centro de Química, Laboratorio de Bioquímica y Biología Molecular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (A.R.-E.); (I.H.-C.); (L.M.-P.-P.)
- Posgrado en Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico
| | - Irma Herrera-Camacho
- Centro de Química, Laboratorio de Bioquímica y Biología Molecular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (A.R.-E.); (I.H.-C.); (L.M.-P.-P.)
| | - Lourdes Millán-Pérez-Peña
- Centro de Química, Laboratorio de Bioquímica y Biología Molecular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico; (A.R.-E.); (I.H.-C.); (L.M.-P.-P.)
| | - Gerardo Santos-López
- Centro de Investigación Biomédica de Oriente, Laboratorio de Biología Molecular y Virología, Instituto Mexicano del Seguro Social (IMSS), Metepec 74360, Mexico;
| | - José F. Rivera-Benítez
- Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Ciudad de México 38110, Mexico;
| |
Collapse
|
26
|
Sharma A, Sharma A, Soubani AO. Incidence and inhospital outcomes of coronavirus disease 2019-associated pulmonary aspergillosis in the United States. Ann Thorac Med 2024; 19:87-95. [PMID: 38444990 PMCID: PMC10911242 DOI: 10.4103/atm.atm_190_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 03/07/2024] Open
Abstract
OBJECTIVE The aim of this study was to estimate the predictors, associations, and outcomes of COVID-19-associated pulmonary disease (CAPA) in the United States. STUDY DESIGN AND METHODS This retrospective cohort study was performed by using the National Inpatient Sample Database 2020 to identify coronavirus disease 2019 (COVID-19) and CAPA hospitalizations. Baseline variables and outcomes were compared between COVID-19 hospitalizations without aspergillosis and those with aspergillosis. These variables were then used to perform an adjusted analysis for obtaining predictors and factors associated with CAPA and its inhospital mortality. RESULTS Of the 1,020,880 hospitalizations identified with the principal diagnosis of COVID-19, CAPA was identified in 1510 (0.1%) hospitalizations. The CAPA cohort consisted of a higher proportion of males (58%) as well as racial and ethnic minorities (Hispanics, Blacks, and others [including Asian or Pacific islanders, native Americans]). Inhospital mortality was significantly higher (47.35% vs. 10.87%, P < 0.001), the average length of stay was longer (27.61 vs. 7.29 days, P < 0.001), and the mean cost per hospitalization was higher ($121,560 vs. $18,423, P < 0.001) in the CAPA group compared to COVID-19 without aspergillosis. History of solid organ transplant, chronic obstructive pulmonary disease, and venous thromboembolism were associated with higher odds of CAPA among other factors. The use of invasive mechanical ventilation (adjusted odds ratio [aOR] 6.24, P < 0.001), acute kidney injury (aOR 2.02, P = 0.028), and septic shock (aOR 2.07, P = 0.018) were associated with higher inhospital mortality in the CAPA cohort. CONCLUSION While CAPA is an infrequent complication during hospitalizations for COVID-19, it significantly increases all-cause mortality, prolongs hospital stays, and leads to higher hospital expenses compared to COVID-19 cases without aspergillosis.
Collapse
Affiliation(s)
- Aditya Sharma
- Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Aditi Sharma
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ayman O. Soubani
- Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
27
|
Ramos-Usuga D, Jimenez-Marin A, Cabrera-Zubizarreta A, Benito-Sanchez I, Rivera D, Martínez-Gutiérrez E, Panera E, Boado V, Labayen F, Cortes JM, Arango-Lasprilla JC. Cognitive and brain connectivity trajectories in critically ill COVID-19 patients. NeuroRehabilitation 2024; 54:359-371. [PMID: 38393927 DOI: 10.3233/nre-230216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
BACKGROUND Multiple Organ failure (MOF) is one of the main causes of admission to the Intensive Care Unit (ICU) of patients infected with COVID-19 and can cause short- and long-term neurological deficits. OBJECTIVE To compare the cognitive functioning and functional brain connectivity at 6-12 months after discharge in two groups of individuals with MOF, one due to COVID-19 and the other due to another cause (MOF-group), with a group of Healthy Controls (HC). METHODS Thirty-six participants, 12 from each group, underwent a neuropsychological and neuroimaging assessment at both time-points. Functional connectivity of the resting state networks was compared between COVID-19 and HC while controlling for the effect of MOF. The association between functional connectivity and neuropsychological performance was also investigated. RESULTS Compared to the HC, COVID-19 group demonstrated hypoconnectivity between the Default Mode Network and Salience Network. This pattern was associated with worse performance on tests of attention and information processing speed, at both time-points. CONCLUSION The study of the association between cognitive function and brain functional connectivity in COVID-19 allows the understanding of the short- and long-term neurological alterations of this disease and promotes the development of intervention programs to improve the quality of life for this understudied population.
Collapse
Affiliation(s)
- Daniela Ramos-Usuga
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Biomedical Research Doctorate Program, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Antonio Jimenez-Marin
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Biomedical Research Doctorate Program, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | - Itziar Benito-Sanchez
- Biomedical Research Doctorate Program, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Diego Rivera
- Department of Health Sciences, Public University of Navarre, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Endika Martínez-Gutiérrez
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Dipartamento Interateneo di Fisica, National Institute for Nuclear Physics - Bari, Bari, Italy
| | - Elena Panera
- Intensive Care Unit, Cruces University Hospital, Barakaldo, Spain
| | - Victoria Boado
- Intensive Care Unit, Cruces University Hospital, Barakaldo, Spain
| | - Fermín Labayen
- Intensive Care Unit, Cruces University Hospital, Barakaldo, Spain
| | - Jesus M Cortes
- Biobizkaia Health Research Institute, Barakaldo, Spain
- IKERBASQUE, The Basque Foundation for Science, Bilbao, Spain
- Department of Cell Biology and Histology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | |
Collapse
|
28
|
Raj K, Kumari B, Kumar S, Bansal A, Mohanraj PS, Dey S, Thakur A. The Prognostic Value of De Ritis Ratio on the Survival Rate of ICU-Admitted COVID-19 Patients During the Second Wave: A Retrospective Study in a Tertiary Care Hospital in India. Cureus 2024; 16:e52484. [PMID: 38371091 PMCID: PMC10873898 DOI: 10.7759/cureus.52484] [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] [Accepted: 01/18/2024] [Indexed: 02/20/2024] Open
Abstract
Background Prognostic markers are essential for optimizing COVID-19 patient care. This retrospective study examines the prognostic value of the De Ritis ratio (DRR) in intensive care unit (ICU)-admitted patients during the second wave of the pandemic. Methods A retrospective study of four-month duration (March to June 2021) was conducted on 161 ICU-admitted COVID-19 patients in a tertiary care hospital in India. The data included demographics, comorbidities, laboratory results, ICU admission dates, and survival outcomes. The De Ritis ratio was calculated on day 0, day 2, and day 5. The analyses included descriptive statistics, diagnostic accuracy, and logistic regression. Results Survival rates decreased with ICU stay: day 0 (survival, 58.4%; mortality, 41.6%), day 2 (survival, 54.5%; mortality, 45.5%), and day 5 (survival, 49.5%; mortality, 50.5%). De Ritis ratio's diagnostic accuracy varied, with increasing specificity and negative predictive value (NPV). Logistic regression showed higher day 5 De Ritis ratios, and male gender was associated with reduced survival odds. Conclusion The De Ritis ratio demonstrates promise as an early prognostic marker for COVID-19 patients, with an increase in predictive accuracy over time. The results emphasize the De Ritis ratio's potential as an early indicator of disease severity, offering clinicians a tool to recognize patients at higher risk and enhance the effectiveness of critical care interventions.
Collapse
Affiliation(s)
- Khushboo Raj
- Biochemistry, All India Institute of Medical Sciences, Patna, IND
| | - Bandana Kumari
- Biochemistry, All India Institute of Medical Sciences, Patna, IND
| | - Sushil Kumar
- Biochemistry, All India Institute of Medical Sciences, Patna, IND
| | - Akash Bansal
- Biochemistry, All India Institute of Medical Sciences, Gorakhpur, IND
| | | | - Soma Dey
- Biochemistry, All India Institute of Medical Sciences, Patna, IND
| | - Aishwarya Thakur
- Biochemistry, All India Institute of Medical Sciences, Patna, IND
| |
Collapse
|
29
|
Nanavaty D, Sinha R, Kaul D, Sanghvi A, Kumar V, Vachhani B, Singh S, Devarakonda P, Reddy S, Verghese D. Impact of COVID-19 on Acute Myocardial Infarction: A National Inpatient Sample Analysis. Curr Probl Cardiol 2024; 49:102030. [PMID: 37573898 DOI: 10.1016/j.cpcardiol.2023.102030] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023]
Abstract
COVID-19 has been associated with a higher incidence of acute myocardial infarction and related complications. We sought to assess the impact of COVID-19 diagnosis on hospitalizations with an index admission of AMI. The National inpatient sample 2020 was queried for hospitalizations with an index admission of AMI, further stratified for admissions with and without COVID-19. The 2 groups' mortality, procedure, and complication rates were compared using suitable statistical tests. Multivariate regression analysis was further performed to study the impact of COVID-19 on mortality as the primary outcome and length of stay and total hospital cost as secondary outcomes. A total of 555,540 admissions for AMI were identified, of which 5818 (1.04%) had concomitant COVID-19. Hospitalizations in the COVID-19 cohort of both groups had a lower procedure rate for coronary angiography. Thrombolysis use was higher in the STEMI patients with COVID-19. Most cardiac complications in AMI patients were higher when infected with SARS-CoV-2. Multivariate regression analysis revealed that COVID-19 led to higher odds of mortality and total length of stay in AMI hospitalizations. COVID-19 portends a worse prognosis in hospitalizations with AMI. These admissions have a significantly higher mortality rate and increased complications.
Collapse
Affiliation(s)
- Dhairya Nanavaty
- Department of Internal Medicine, The Brooklyn Hospital Center, NY.
| | - Rishav Sinha
- Department of Internal Medicine, The Brooklyn Hospital Center, NY
| | - Diksha Kaul
- Department of Internal Medicine, The Brooklyn Hospital Center, NY
| | - Ankushi Sanghvi
- Department of Internal Medicine, Saint Vincent Hospital, Worcester, MA
| | - Vikash Kumar
- Department of Internal Medicine, The Brooklyn Hospital Center, NY
| | | | - Sohrab Singh
- Department of Cardiology, The Brooklyn Hospital Center, NY
| | | | - Sarath Reddy
- Department of Cardiology, The Brooklyn Hospital Center, NY
| | | |
Collapse
|
30
|
Georgakopoulou VE, Gkoufa A, Makrodimitri S, Tsakanikas A, Basoulis D, Voutsinas PM, Karamanakos G, Eliadi I, Samara S, Triantafyllou M, Eleftheriadou I, Kampouropoulou O, Papageorgiou CV, Anastasopoulou A, Papalexis P, Trakas I, Trakas N, Spandidos DA, Steiropoulos P, Sipsas NV. Risk factors for the in‑hospital and 1‑year mortality of elderly patients hospitalized due to COVID‑19‑related pneumonia. Exp Ther Med 2024; 27:22. [PMID: 38125348 PMCID: PMC10728907 DOI: 10.3892/etm.2023.12310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/09/2023] [Indexed: 12/23/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is characterized by poor outcomes and a high mortality rate, particularly among elderly patients. Since the beginning of the pandemic, an older age has been recognized as a critical risk factor for disease severity, with increasing mortality rates in each decade of life. This phenomenon may be a consequence of a poor previous health status, with a higher prevalence of pre-existing comorbidities and a higher degree of frailty. The majority of studies on the outcomes and risk factors of elderly patients refer to the first waves of the pandemic and the predictors of in-hospital mortality in these patients. The aim of the present study was to provide a detailed description of the clinical characteristics and management of a cohort of elderly patients (≥65 years of age) who were hospitalized with COVID-19-related pneumonia in all phases of the pandemic, presenting their outcomes, and investigating predictors of in-hospital and out-of-hospital mortality over a period of 1 year in this particularly vulnerable population. A total of 1,124 elderly patients (603 males, 53.7%) with a mean age of 78.51±7.42 years and a median Charlson comorbidity index (CCI) of 5 were included in the study. Of these patients, 104 (9.3%) were hospitalized during the period of prevalence of the original strain Wuhan, 385 (34.3%) were hospitalized during the period of prevalence of the Alpha variant, 221 (19.7%) were hospitalized during the period of prevalence of the Delta variant, and 414 (36.8%) were hospitalized during the period of prevalence of the Omicron variant. Overall, the in-hospital mortality rate was 33.4% (375 patients), and the 1-year mortality rate was 44.7% (502 patients). The majority of patients had not been vaccinated or had not completed full vaccination against severe acute respiratory syndrome coronavirus-2 (843 patients, 75%), given the period of infection. Age, immature granulocytes, lactate dehydrogenase (LDH) levels, ferritin levels, chest X-ray score, as well as the absence of full vaccination, cough and fatigue, were statistically significantly and independently associated with in-hospital mortality, while age, LDH levels, ferritin levels, alanine aminotransferase levels, CCI, chest X-ray score, the absence of cough and fatigue, and a history of dementia were statistically significantly and independently associated with 1-year mortality. On the whole, the present study demonstrates that both the in-hospital mortality and 1-year mortality rates of elderly patients hospitalized due to COVID-19-related pneumonia are high.
Collapse
Affiliation(s)
- Vasiliki Epameinondas Georgakopoulou
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Pathophysiology, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Aikaterini Gkoufa
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Sotiria Makrodimitri
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Aristeidis Tsakanikas
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios Basoulis
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Pathophysiology, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Pantazis M. Voutsinas
- Department of Pathophysiology, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgios Karamanakos
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Irene Eliadi
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Stamatia Samara
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Triantafyllou
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioanna Eleftheriadou
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Olga Kampouropoulou
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Chrysovalantis V. Papageorgiou
- Pulmonology Department, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Amalia Anastasopoulou
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Petros Papalexis
- Unit of Endocrinology, First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
| | - Ilias Trakas
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Trakas
- Department of Biochemistry, Sismanogleio Hospital, 15126 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Paschalis Steiropoulos
- Department of Pulmonology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Nikolaos V. Sipsas
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Pathophysiology, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
31
|
Assiri AM, Alamaa T, Elenezi F, Alsagheir A, Alzubaidi L, TIeyjeh I, Alhomod AS, Gaffas EM, Amer SA. Unveiling the Clinical Spectrum of Post-COVID-19 Conditions: Assessment and Recommended Strategies. Cureus 2024; 16:e52827. [PMID: 38406111 PMCID: PMC10884364 DOI: 10.7759/cureus.52827] [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] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
SARS-CoV-2 caused the pandemic of the rapidly evolving COVID-19. As of December 6, 2023, there were 765,152,854 COVID-19-recovering cases. Long-term consequences known as "long COVID" and "post-COVID-19 conditions" (PCCs) or "post-acute COVID-19 syndrome" are being reported more frequently in a subset of recovering patients. Systemic, neuropsychiatric, cardio-respiratory, and gastrointestinal symptoms are the most prevalent. The management of PCCs poses unique challenges due to the lack of official guidelines and the complex nature of the illness. This abstract highlights key principles derived from recent reviews and expert recommendations to provide healthcare professionals with a comprehensive approach to manage post-COVID-19 patients. Preventive medicine plays a crucial role in managing PCCs. While no specific medications are available for treatment, preventive measures such as COVID-19 vaccination, adherence to precautionary measures, regular consultations with medical professionals, monitoring symptoms and progress, and seeking information on symptom management are essential to assist patients in their recovery and improve their quality of life. Medical management requires transparent goal-setting and collaborative decision-making based on the patient's symptoms, comorbidities, and treatment objectives. Treatment plans for post-COVID-19 patients should focus on patient education, using registries and calendars to track symptoms and triggers, providing support and reassurance, and offering holistic support through peer networks and supportive psychotherapy techniques. Symptomatic and rehabilitative care, including well-established symptom management techniques, physical rehabilitation programs, and addressing mental health and well-being, are vital components of post-COVID-19 management. Lifestyle factors such as stress reduction, nutrition, and sleep should be incorporated into managing underlying medical conditions in post-COVID-19 patients. Regular follow-up visits and referrals to specialists are recommended to monitor the patient's progress and address specific organ system involvement or additional care needs. In summary, for the effective management of PCCs, a holistic approach should include preventive measures, patient education, supportive psychotherapy, symptomatic and rehabilitative care, medical management, counseling on lifestyle elements, and appropriate follow-up plans. However, it is crucial to stay updated with evolving guidelines and recommendations from healthcare authorities to provide the most effective and evidence-based care to post-COVID-19 patients.
Collapse
Affiliation(s)
| | - Tareef Alamaa
- Therapeutic Services, Saudi Ministry of Health, Riyadh, SAU
| | - Faisal Elenezi
- Assistant Agency for Hospital Affairs, Saudi Ministry of Health, Riyadh, SAU
| | - Aeshah Alsagheir
- Health Quality Index Measuring, Saudi Ministry of Health, Riyadh, SAU
| | - Lamya Alzubaidi
- Assistant Agency for Hospital Affairs, Saudi Ministry of Health, Riyadh, SAU
| | - Imad TIeyjeh
- Infectious Diseases, Mayo Clinic, Rochester, USA
- Infectious Diseases, King Fahad Medical City, Riyadh, SAU
| | | | - Eisha M Gaffas
- Mental Health and Social Services, Saudi Ministry of Health, Riyadh, SAU
| | - Samar A Amer
- Public Health and Community Medicine, Zagazig University, Zagazig, EGY
- General Administration of Health Programs and Non-communicable Diseases, Saudi Ministry of Health, Riyadh, SAU
| |
Collapse
|
32
|
Krishna N, K P S, G K R. Identifying diseases associated with Post-COVID syndrome through an integrated network biology approach. J Biomol Struct Dyn 2024; 42:652-671. [PMID: 36995291 DOI: 10.1080/07391102.2023.2195003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023]
Abstract
A growing body of research shows that COVID-19 is now recognized as a multi-organ disease with a wide range of manifestations that can have long-lasting repercussions, referred to as post-COVID-19 syndrome. It is unknown why the vast majority of COVID-19 patients develop post-COVID-19 syndrome, or why patients with pre-existing disorders are more likely to experience severe COVID-19. This study used an integrated network biology approach to obtain a comprehensive understanding of the relationship between COVID-19 and other disorders. The approach involved building a PPI network with COVID-19 genes and identifying highly interconnected regions. The molecular information contained within these subnetworks, as well as the pathway annotations, were used to reveal the link between COVID-19 and other disorders. Using Fisher's exact test and disease-specific gene information, significant COVID-19-disease associations were discovered. The study discovered diseases that affect multiple organs and organ systems, thus proving the theory of multiple organ damage caused by COVID-19. Cancers, neurological disorders, hepatic diseases, cardiac disorders, pulmonary diseases, and hypertensive diseases are just a few of the conditions linked to COVID-19. Pathway enrichment analysis of shared proteins revealed the shared molecular mechanism of COVID-19 and these diseases. The findings of the study shed new light on the major COVID-19-associated disease conditions and how their molecular mechanisms interact with COVID-19. The novelty of studying disease associations in the context of COVID-19 provides new insights into the management of rapidly evolving long-COVID and post-COVID syndromes, which have significant global implications.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Navami Krishna
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Sijina K P
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Rajanikant G K
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| |
Collapse
|
33
|
Strubchevska K, Rachkovska A, Krenytska D, Karbovskyy V, Kozyk M, Secor B, Raksha N, Vovk T, Savchuk O, Falalyeyeva T, Kaminsky R, Ostapchenko L. Coagulation Parameters in Post-Covid-19 Condition in Relation to Various Titers of Anti-SARS-CoV-2 IgG in Blood Plasma. Int J Gen Med 2023; 16:6127-6135. [PMID: 38156079 PMCID: PMC10754419 DOI: 10.2147/ijgm.s425496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023] Open
Abstract
Background and Objectives Post-COVID-19 condition is thought to affect 10-20% of people at least 3 months after a diagnosis of COVID-19 and two months of symptoms. Post-COVID-19 condition presents itself with many clinical effects with varying degrees of severity ranging from a mild cough to a life-threatening coagulopathy. Our study aimed to identify a relationship between the titers of anti-SARS-CoV-2 IgG and anticoagulation parameters: antithrombin III (ATIII), protein C (PC) and thrombomodulin (TM). Materials and Methods Blood plasma was collected from healthy donors aged 25-45 who had recovered from COVID-19 3-6 months ago and their titers of anti-SARS-CoV-2 IgG and ATIII, PC, and TM were measured. Results We found that concentrations and activities of key anticoagulation parameters (ATIII, PC, and TM) measured in donor plasma during the post-COVID-19 varied in relation to the titers of anti-SARS-CoV-2 IgG. Conclusion While we identified a dysfunction of anticoagulation parameters in patients with post-COVID-19, we aim to explore the subpopulation antibody IgG fraction directly using in vivo and in vitro experiments with the possibility to contribute to the development of treatment options for post-COVID-19 conditions.
Collapse
Affiliation(s)
- Kateryna Strubchevska
- Department of Internal Medicine, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Antonina Rachkovska
- Department of Internal Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Daryna Krenytska
- Department of Internal Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | | | - Marko Kozyk
- Department of Internal Medicine, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Benjamin Secor
- Department of Internal Medicine, Oakland University William Beaumont School of Medicine, Auburn Hills, MI, USA
| | - Nataliia Raksha
- Department of Internal Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetiana Vovk
- Department of Internal Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olexii Savchuk
- Department of Internal Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetyana Falalyeyeva
- Department of Internal Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Rostyslav Kaminsky
- Department Educational-Scientific Center ”Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Liudmyla Ostapchenko
- Department of Internal Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| |
Collapse
|
34
|
Zisis SN, Durieux JC, Mouchati C, Funderburg N, Ailstock K, Chong M, Labbato D, McComsey GA. Arterial Stiffness and Oxidized LDL Independently Associated With Post-Acute Sequalae of SARS-CoV-2. Pathog Immun 2023; 8:1-15. [PMID: 38156116 PMCID: PMC10753933 DOI: 10.20411/pai.v8i2.634] [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: 09/28/2023] [Accepted: 11/11/2023] [Indexed: 12/30/2023] Open
Abstract
OBJECTIVE COVID-19 survivors can experience lingering symptoms known as post-acute sequelae of SARS-CoV-2 (PASC) that appear in different phenotypes, and its etiology remains elusive. We assessed the relationship of endothelial dysfunction with having COVID and PASC. METHODS Data was collected from a prospectively enrolled cohort (n=379) of COVID-negative and COVID-positive participants with and without PASC. Primary outcomes, endothelial function (measured by reactive hyperemic index [RHI]), and arterial elasticity (measured by augmentation index standardized at 75 bpm [AI]), were measured using the FDA approved EndoPAT. Patient characteristics, labs, metabolic measures, markers of inflammation, and oxidized LDL (ox-LDL) were collected at each study visit, and PASC symptoms were categorized into 3 non-exclusive phenotypes: cardiopulmonary, neurocognitive, and general. COVID-negative controls were propensity score matched to COVID-negative-infected cases using the greedy nearest neighbor method. RESULTS There were 14.3% of participants who were fully recovered COVID positive and 28.5% who were COVID positive with PASC, averaging 8.64 ± 6.26 total number of symptoms. The mean RHI was similar across the cohort and having COVID or PASC was not associated with endothelial function (P=0.33). Age (P<0.0001), female sex (P<0.0001), and CRP P=0.04) were positively associated with arterial stiffness, and COVID positive PASC positive with neurological and/or cardiopulmonary phenotypes had the worst arterial elasticity (highest AI). Values for AI (P=0.002) and ox-LDL (P<0.0001) were independently and positively associated with an increased likelihood of having PASC. CONCLUSION There is evidence of an independent association between PASC, ox-LDL, and arterial stiffness with neurological and/or cardiopulmonary phenotypes having the worst arterial elasticity. Future studies should continue investigating the role of oxidative stress in the pathophysiology of PASC.
Collapse
Affiliation(s)
- Sokratis N Zisis
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jared C Durieux
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Nicholas Funderburg
- School of Health and Rehabilitation Sciences, Ohio State University, Columbus, Ohio
| | - Kate Ailstock
- School of Health and Rehabilitation Sciences, Ohio State University, Columbus, Ohio
| | - Mary Chong
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Danielle Labbato
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Grace A McComsey
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
35
|
Kahaleh AA, Ellington TM, Coker AO, Tillery E, Manasco KB, Truong HA, Kebodeaux CD, Edwards KL, Hayatshahi A, Gumina G, Hughes JA, Fuentes DG. Impact of COVID-19 on Pharmacy Education and Practice: Strategies to Boost Advocacy and Unity among Health Care Organizations. AMERICAN JOURNAL OF PHARMACEUTICAL EDUCATION 2023; 87:100091. [PMID: 37953084 DOI: 10.1016/j.ajpe.2023.100091] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 11/14/2023]
Abstract
The global COVID-19 pandemic impacted pharmacy education and changed the pharmacists' scope of practice at the federal and state levels. Based on the Amended Public Readiness and Emergency Preparedness Act, pharmacists were authorized to provide essential services, including testing, treatments, and immunizations at various practice settings. Specifically, the United States Food and Drug Administration issued emergency use authorization for several medications, vaccines, and medical devices. The pandemic also affected the regulatory landscape for pharmacists, pharmacy education, access to care, and delivery of pharmacy services in-person and through telehealth. The pandemic's specific impact on pharmacy education heightened awareness of the well-being of the Academy. This commentary will highlight the impact of COVID-19 on both pharmacy education and practice. It will also provide strategies that educators, researchers, and practitioners can take into future research and action to help promote advocacy and unity among pharmacy organizations.
Collapse
Affiliation(s)
- Abby A Kahaleh
- Roosevelt University College of Science, Health, & Pharmacy, Schaumburg, IL, USA.
| | - Thomas M Ellington
- Bernard J. Dunn School of Pharmacy, Shenandoah University, Winchester, VA, USA
| | - Adeola O Coker
- University of the Incarnate Word Feik School of Pharmacy, San Antonio, TX, USA
| | - Erika Tillery
- Presbyterian College School of Pharmacy, Clinton, SC, USA
| | - Kalen B Manasco
- University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Hoai-An Truong
- University of Maryland Eastern Shore, Princess Anne, MD, USA
| | | | - Krystal L Edwards
- Texas Tech University Health Sciences Center Jerry H. Hodge School of Pharmacy, Dallas, TX, USA
| | | | | | - Jeremy A Hughes
- Chicago State University College of Pharmacy, Chicago, IL, USA
| | - David G Fuentes
- University of Portland School of Nursing and Health Innovations, Portland, OR, USA; American Journal of Pharmaceutical Education, USA
| |
Collapse
|
36
|
Paul I, Roy A, Ray S. Molecular Design of Novel Inhibitor by Targeting IL-6Rα using Combined Pharmacophore and Experimentally Verified Plant Products with Scaffold-Hopping Techniques: A Dual Therapeutic Strategy for COVID-19 and Cancer. Chem Biodivers 2023; 20:e202300806. [PMID: 37967248 DOI: 10.1002/cbdv.202300806] [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: 06/02/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/17/2023]
Abstract
The IL-6/IL-6R/gp130 complex serves as a significant indicator of cytokine release syndrome in COVID-19 and chronic inflammation, increasing the risk of cancer. Therefore, we identified IL-6Rα as a potential target to block gp130 interaction. Notably, there has been no reception of approval for an orally available drug to serve this purpose, to date. In this study, we targeted IL-6Rα to inhibit IL-6Rα/gp130 interaction. The selection of the lead candidate L821 involved the amalgamation of three drug discovery approaches. This library was screened employing tertiary structure-based pharmacophore models followed by molecular docking models, scaffold-hopping, MM/PBSA as well as MM/GBSA analysis, and assessments of pKi and ADMET properties. After evaluating the binding interactions with key amino acids, 15 potential ligands were chosen, with the top ligand undergoing further investigation by means of molecular dynamics simulations. Considering the stability of the complexes, the strong interactions observed between ligand and residues of IL-6Rα/gp130, and the favorable binding free energy calculations, L821 emerged as the prime candidate for inhibiting IL-6Rα. Notably, L821 exhibited a docking-based binding affinity of -9.5 kcal/mol. Our study presents L821 as a promising inhibitor for future in vitro analysis, potentially combatting SARS-CoV-2-related cytokine storms and serving as an oncogenic drug therapy.
Collapse
Affiliation(s)
- Ishani Paul
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Alankar Roy
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Sujay Ray
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| |
Collapse
|
37
|
Pooladgar P, Sakhabakhsh M, Soleiman-Meigooni S, Taghva A, Nasiri M, Darazam IA. The effect of donepezil hydrochloride on post-COVID memory impairment: A randomized controlled trial. J Clin Neurosci 2023; 118:168-174. [PMID: 37952347 DOI: 10.1016/j.jocn.2023.09.005] [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: 04/27/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Post-Coronavirus Disease (COVID-19) condition, known as "post-COVID syndrome," is associated with a range of complications persisting even after recovery. Among these complications, cognitive dysfunction, including memory impairment, has been relatively common observed, impacting executive function and quality of life. To date, no approved treatment exists for this specific complication. Therefore, the present clinical trial aimed to investigate the impact of Donepezil Hydrochloride on post-COVID memory impairment. METHODS A randomized, controlled trial (Approval ID: IRCT20210816052203N1) was conducted, enrolling 25 patients with post-COVID memory impairment. Participants with a history of hospitalization were randomly assigned to either the drug group (n = 10) or the control group (n = 15). Memory indices were assessed at baseline, one month, and three months later using the Wechsler Memory Scale-Revised test. SPSS software and appropriate statistical tests were employed for data analysis. RESULTS The statistical analysis revealed no significant difference in WMS-R subtest and index scores between the drug and control groups at the 4-week and 12-week follow-up periods. However, within the drug group, there was a notable increase in the visual reproduction I and verbal paired associates II subtests during the specified time intervals. CONCLUSION While donepezil 5 mg did not exhibit a significant overall increase in memory scales compared to the control group over time, our findings suggest that this medication may exert a positive effect on specific memory subtests. Further research and exploration are warranted to better understand the potential benefits of donepezil in managing post-COVID-related memory impairment. TRIAL REGISTRATION The study was approved by the Research Ethics Committee of Aja University of Medical Sciences (Approval ID: IR.AJAUMS.REC.1400.125) and registered in the Iranian Registry of Clinical Trials (IRCT) (Approval ID: IRCT20210816052203N1).
Collapse
Affiliation(s)
- Parham Pooladgar
- Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran; School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mehdi Sakhabakhsh
- Head of Department of Neurology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran.
| | | | - Arsia Taghva
- Cognitive Science and Behavioral Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Malihe Nasiri
- Basic Science Department, School of Nursing & Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ilad Alavi Darazam
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Indriani S, Amshar M, Nugraha RA, Siddiq T, Adiarto S. Acute Limb Ischemia in COVID-19: A Systematic Review. Int J Angiol 2023; 32:215-226. [PMID: 37927841 PMCID: PMC10624540 DOI: 10.1055/s-0043-1771250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Abstract
Numerous studies have reported cases of acute limb ischemia (ALI) in coronavirus disease 2019 (COVID-19), along with a sharp increase of ALI incidence during the COVID-19 pandemic. The objective of this study was to analyze the clinical features, treatment strategies, and outcomes in COVID-19 patients who developed ALI. A systematic search was performed in PubMed, ScienceDirect, and Cochrane to identify relevant articles. Variables of interest included ALI location, ALI and COVID-19 severity, concomitant thrombosis, comorbidities/risk factors, coagulation and inflammatory parameters, therapy, outcome, and cause of death. Data extraction and pooling were conducted by two reviewers. Fifty-six articles with a total of 191 subjects were included in the analysis; 78.5% of the subjects were male; mean age was 64.2 years; 94.2% of cases were ALI de novo; 41.1% of the subjects had ALI stage IIB; 82.7% of ALI occurred in the lower limb. The popliteal artery was the most commonly affected artery (48.2% of subjects); 38.1% of the subjects had severe COVID-19; 11.1% had asymptomatic COVID-19, and 15.9% were post-COVID-19; 50.7% of the subjects had hypertension; 12.0% of the subjects had concomitant thrombosis; 98.1% of the subjects had elevated D-dimer, and 83.3% of the subjects had elevated C-reactive protein; 61.9% of the subjects underwent surgical thromboembolectomy, 21.3% underwent conservative treatment, and 8.1% underwent amputation initially. The overall mortality rate and limb amputation rate were 39.3 and 22.4%, respectively; 67.6% of deaths were due to respiratory failure. Our study supports that COVID-19 is associated with hypercoagulability that may trigger the development of ALI, and that ALI is a possible predictor of poor outcome in COVID-19 patients.
Collapse
Affiliation(s)
- Suci Indriani
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Division of Vascular Medicine, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Mohamed Amshar
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Raka Aldy Nugraha
- Department of General Medicine, Universitas Indonesia Hospital, Depok, West Java, Indonesia
| | - Taofan Siddiq
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Division of Vascular Medicine, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Suko Adiarto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Division of Vascular Medicine, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| |
Collapse
|
39
|
Schmidt M, Hébert S, Wallukat G, Ponader R, Krickau T, Galiano M, Reutter H, Woelfle J, Agaimy A, Mardin C, Hoerning A, Hohberger B. "Multisystem Inflammatory Syndrome in Children"-Like Disease after COVID-19 Vaccination (MIS-V) with Potential Significance of Functional Active Autoantibodies Targeting G-Protein-Coupled Receptors (GPCR-fAAb) for Pathophysiology and Therapy. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1836. [PMID: 38136038 PMCID: PMC10741397 DOI: 10.3390/children10121836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/28/2023] [Accepted: 10/28/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND An infection with SARS-CoV-2 can trigger a systemic disorder by pathological autoimmune processes. A certain type of this dysregulation is known as Multisystemic inflammatory syndrome in children (MIS-C). However, similar symptoms may occur and have been described as Multisystemic inflammatory syndrome after SARS-CoV-2 Vaccination (MIS-V) following vaccination against SARS-CoV-2. We report the case of a 12-year-old boy who was identified with MIS-C symptoms without previous SARS-CoV-2 infection after receiving two doses of the Pfizer-BioNTech COVID-19 vaccine approximately one month prior to the onset of symptoms. He showed polyserositis, severe gastrointestinal symptoms and, consequently, a manifestation of a multiorgan failure. IgG antibodies against spike proteins of SARS-CoV-2 were detected, indicating a successful vaccination, while SARS-CoV-2 Nucleocapsid protein antibodies and SARS-CoV-2 PCR were not detected. Several functional, active autoantibodies against G-protein-coupled receptors (GPCR-fAAb), previously associated with Long COVID disease, were detected in a cardiomyocyte bioassay. Immunosuppression with steroids was initiated. Due to side effects, treatment with steroids and later interleukin 1 receptor antagonists had to be terminated. Instead, immunoadsorption was performed and continued with tacrolimus and mycophenolic acid therapy, leading to improvement and discharge after 79 days. GPCR-fAAb decreased during therapy and remained negative after clinical curing and under continued immunosuppressive therapy with tacrolimus and mycophenolic acid. Follow-up of the patient showed him in good condition after one year. CONCLUSIONS Infection with SARS-CoV-2 shows a broad and severe variety of symptoms, partly due to autoimmune dysregulation, which, in some instances, can lead to multiorgan failure. Despite its rarity, post-vaccine MIS-C-like disease may develop into a serious condition triggered by autoimmune dysregulation. The evidence of circulating GPCR-fAAb and their disappearance after therapy suggests a link of GPCR-fAAb to the clinical manifestations. Thus, we hypothesize a potential role of GPCR-fAAb in pathophysiology and their potential importance for the therapy of MIS-C or MIS-V. However, this observation needs further investigation to prove a causative correlation.
Collapse
Affiliation(s)
- Marius Schmidt
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | - Steven Hébert
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | | | - Rolf Ponader
- Department of Pediatrics and Adolescent Medicine, 95032 Hof, Germany
| | - Tobias Krickau
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | - Matthias Galiano
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | - Heiko Reutter
- Division of Neonatology and Pediatric Intensive Care, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
| | - Abbas Agaimy
- Department of Pathology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Christian Mardin
- Department of Ophthalmology, University Hospital Erlangen, 90766 Erlangen, Germany
| | - André Hoerning
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany (A.H.)
- German Center for Immunotherapy, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, University Hospital Erlangen, 90766 Erlangen, Germany
- German Center for Immunotherapy, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, 91054 Erlangen, Germany
| |
Collapse
|
40
|
Stadler JT, Habisch H, Prüller F, Mangge H, Bärnthaler T, Kargl J, Pammer A, Holzer M, Meissl S, Rani A, Madl T, Marsche G. HDL-Related Parameters and COVID-19 Mortality: The Importance of HDL Function. Antioxidants (Basel) 2023; 12:2009. [PMID: 38001862 PMCID: PMC10669705 DOI: 10.3390/antiox12112009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
COVID-19, caused by the SARS-CoV-2 coronavirus, emerged as a global pandemic in late 2019, resulting in significant global public health challenges. The emerging evidence suggests that diminished high-density lipoprotein (HDL) cholesterol levels are associated with the severity of COVID-19, beyond inflammation and oxidative stress. Here, we used nuclear magnetic resonance spectroscopy to compare the lipoprotein and metabolic profiles of COVID-19-infected patients with non-COVID-19 pneumonia. We compared the control group and the COVID-19 group using inflammatory markers to ensure that the differences in lipoprotein levels were due to COVID-19 infection. Our analyses revealed supramolecular phospholipid composite (SPC), phenylalanine, and HDL-related parameters as key discriminators between COVID-19-positive and non-COVID-19 pneumonia patients. More specifically, the levels of HDL parameters, including apolipoprotein A-I (ApoA-I), ApoA-II, HDL cholesterol, and HDL phospholipids, were significantly different. These findings underscore the potential impact of HDL-related factors in patients with COVID-19. Significantly, among the HDL-related metrics, the cholesterol efflux capacity (CEC) displayed the strongest negative association with COVID-19 mortality. CEC is a measure of how well HDL removes cholesterol from cells, which may affect the way SARS-CoV-2 enters cells. In summary, this study validates previously established markers of COVID-19 infection and further highlights the potential significance of HDL functionality in the context of COVID-19 mortality.
Collapse
Affiliation(s)
- Julia T. Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (T.B.); (J.K.); (A.P.); (M.H.); (S.M.); (A.R.)
| | - Hansjörg Habisch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (H.H.); (T.M.)
| | - Florian Prüller
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria;
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria;
| | - Thomas Bärnthaler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (T.B.); (J.K.); (A.P.); (M.H.); (S.M.); (A.R.)
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (T.B.); (J.K.); (A.P.); (M.H.); (S.M.); (A.R.)
- BioTechMed Graz, 8010 Graz, Austria
| | - Anja Pammer
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (T.B.); (J.K.); (A.P.); (M.H.); (S.M.); (A.R.)
| | - Michael Holzer
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (T.B.); (J.K.); (A.P.); (M.H.); (S.M.); (A.R.)
| | - Sabine Meissl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (T.B.); (J.K.); (A.P.); (M.H.); (S.M.); (A.R.)
| | - Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (T.B.); (J.K.); (A.P.); (M.H.); (S.M.); (A.R.)
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (H.H.); (T.M.)
- BioTechMed Graz, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria; (J.T.S.); (T.B.); (J.K.); (A.P.); (M.H.); (S.M.); (A.R.)
- BioTechMed Graz, 8010 Graz, Austria
| |
Collapse
|
41
|
Flynn J, Ahmadi MM, McFarland CT, Kubal MD, Taylor MA, Cheng Z, Torchia EC, Edwards MG. Crowdsourcing temporal transcriptomic coronavirus host infection data: Resources, guide, and novel insights. Biol Methods Protoc 2023; 8:bpad033. [PMID: 38107402 PMCID: PMC10723038 DOI: 10.1093/biomethods/bpad033] [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: 08/09/2023] [Revised: 10/07/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
The emergence of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) reawakened the need to rapidly understand the molecular etiologies, pandemic potential, and prospective treatments of infectious agents. The lack of existing data on SARS-CoV-2 hampered early attempts to treat severe forms of coronavirus disease-2019 (COVID-19) during the pandemic. This study coupled existing transcriptomic data from severe acute respiratory syndrome-related coronavirus 1 (SARS-CoV-1) lung infection animal studies with crowdsourcing statistical approaches to derive temporal meta-signatures of host responses during early viral accumulation and subsequent clearance stages. Unsupervised and supervised machine learning approaches identified top dysregulated genes and potential biomarkers (e.g. CXCL10, BEX2, and ADM). Temporal meta-signatures revealed distinct gene expression programs with biological implications to a series of host responses underlying sustained Cxcl10 expression and Stat signaling. Cell cycle switched from G1/G0 phase genes, early in infection, to a G2/M gene signature during late infection that correlated with the enrichment of DNA damage response and repair genes. The SARS-CoV-1 meta-signatures were shown to closely emulate human SARS-CoV-2 host responses from emerging RNAseq, single cell, and proteomics data with early monocyte-macrophage activation followed by lymphocyte proliferation. The circulatory hormone adrenomedullin was observed as maximally elevated in elderly patients who died from COVID-19. Stage-specific correlations to compounds with potential to treat COVID-19 and future coronavirus infections were in part validated by a subset of twenty-four that are in clinical trials to treat COVID-19. This study represents a roadmap to leverage existing data in the public domain to derive novel molecular and biological insights and potential treatments to emerging human pathogens.
Collapse
Affiliation(s)
- James Flynn
- Illumina Corporation, San Diego, CA 92122, United States
| | - Mehdi M Ahmadi
- Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | | | | | - Mark A Taylor
- Bioinfo Solutions LLC, Parker, CO 80134, United States
| | - Zhang Cheng
- Illumina Corporation, San Diego, CA 92122, United States
| | - Enrique C Torchia
- Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | | |
Collapse
|
42
|
Kumar M. Hydrogen sulfide: From a toxic gas to a potential therapy for COVID-19 and inflammatory disorders. Nitric Oxide 2023; 140-141:8-15. [PMID: 37648016 DOI: 10.1016/j.niox.2023.08.002] [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] [Received: 05/15/2023] [Revised: 07/06/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
COVID-19 has been shown to induce inflammatory disorders and CNS manifestations. Swift and efficient treatment strategies are urgently warranted for the management of COVID, inflammatory and neurological disorders. Hydrogen sulfide (H2S) has been associated with several clinical disorders due to its potential to influence a broad range of biological signalling pathways. According to recent clinical studies, COVID patients with lower physiological H2S had higher fatality rates. These findings clearly demonstrate an inverse correlation between H2S levels and the severity of COVID-19. H2S has been proposed as a protective molecule because of its antioxidant, anti-inflammatory, and antiviral properties. Various H2S-releasing prodrugs, hybrids and natural compounds have been tested for their therapeutic efficacy in viral infections and inflammatory disorders. In this review, I am highlighting the rationale for using H2S-based interventions for the management of COVID-19 and post-infection inflammatory disorders including neuroinflammation. I am also proposing therepurposing of existing H2S-releasing prodrugs, developing new NO-H2S-hybrids, targeting H2S metabolic pathways, and using H2S-producing dietary supplements as viable defensive strategies against SARS-CoV-2 infection and COVID-19 pathologies.
Collapse
Affiliation(s)
- Mohit Kumar
- Centre for Excellence in Functional Foods, Food and Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Punjab, 140306, India.
| |
Collapse
|
43
|
Prihatni D, Budianto FC, Andriyoko B, Trisa S. The Correlation Between sP-Selectin and Platelet Count in COVID-19 Patients in Referral Hospital, West Java Indonesia. J Blood Med 2023; 14:555-561. [PMID: 37915624 PMCID: PMC10617529 DOI: 10.2147/jbm.s425667] [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: 06/13/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction sP-selectin is a glycoprotein located in α granules of platelet and endothelial's Weibel Palade body, as expression to platelet activation and endothelial cell stimulation by SARS-CoV-2 binding with ACE2 receptor. Consumptive thrombocytopenia is also related to platelet activation. Elevation of sP-selectin and thrombocytopenia are related to COVID-19 complication and often correlated with severity of COVID-19. Purpose Assess the correlation between sP-selectin and platelet in COVID-19 patients at intensive care and non-intensive care. Patients and Methods The study population was hospitalized COVID-19 patients confirmed by Real-Time PCR that underwent platelet examination within 48 hours upon admission, divided into intensive care and non-intensive care group. sP-selectin examination using ELISA methods. Platelet cell count and sP-selectin divided based on normal reference range. Results The subjects consist of 24 were in intensive care, 25 were in non-intensive care group. A 66.7% of subject in intensive care group has an elevation in sP-selection (>44.0 ng/mL). Thrombocytopenia was significantly correlated with intensive group (r =-0.32, p<0.05). The combination of platelet count <150.000/mm3 and sP-selectin >44.0ng/mL was not correlated with the intensive and non-intensive group. Platelet and sP-selectin were not correlated with each other. Conclusion Thrombocytopenia is able to induce the expression of sP-selectin.
Collapse
Affiliation(s)
- Delita Prihatni
- Clinical Pathology Department, Medical Faculty of Padjadjaran University, Hasan Sadikin Hospital, Bandung, West Java, Indonesia
| | - Frany Charisma Budianto
- Clinical Pathology Department, Medical Faculty of Padjadjaran University, Hasan Sadikin Hospital, Bandung, West Java, Indonesia
| | - Basti Andriyoko
- Clinical Pathology Department, Medical Faculty of Padjadjaran University, Hasan Sadikin Hospital, Bandung, West Java, Indonesia
| | | |
Collapse
|
44
|
Harper TC, Oberlick EM, Smith TJ, Nunes DE, Bray MA, Park S, Driscoll CD, Mowbray SF, Antczak C. GATA1 deletion in human pluripotent stem cells increases differentiation yield and maturity of neutrophils. iScience 2023; 26:107804. [PMID: 37720099 PMCID: PMC10500457 DOI: 10.1016/j.isci.2023.107804] [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: 04/17/2023] [Revised: 08/04/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
Human pluripotent stem cell (hPSC)-derived tissues can be used to model diseases in cell types that are challenging to harvest and study at-scale, such as neutrophils. Neutrophil dysregulation, specifically neutrophil extracellular trap (NET) formation, plays a critical role in the prognosis and progression of multiple diseases, including COVID-19. While hPSCs can generate limitless neutrophils (iNeutrophils) to study these processes, current differentiation protocols generate heterogeneous cultures of granulocytes and precursors. Here, we describe a method to improve iNeutrophil differentiations through the deletion of GATA1. GATA1 knockout (KO) iNeutrophils are nearly identical to primary neutrophils in form and function. Unlike wild-type iNeutrophils, GATA1 KO iNeutrophils generate NETs in response to the physiologic stimulant lipopolysaccharide, suggesting they are a more accurate model when performing NET inhibitor screens. Furthermore, through deletion of CYBB, we demonstrate that GATA1 KO iNeutrophils are a powerful tool in determining involvement of a given protein in NET formation.
Collapse
Affiliation(s)
- Thomas C. Harper
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Elaine M. Oberlick
- Respiratory Disease Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Tomas J. Smith
- Respiratory Disease Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Duncan E. Nunes
- Respiratory Disease Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Mark-Anthony Bray
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Seonmi Park
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Corey D. Driscoll
- Respiratory Disease Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Sarah F. Mowbray
- Respiratory Disease Area, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Christophe Antczak
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| |
Collapse
|
45
|
Mitrofanova L, Makarov I, Goncharova E, Makarova T, Starshinova A, Kudlay D, Shlaykhto E. High Risk of Heart Tumors after COVID-19. Life (Basel) 2023; 13:2087. [PMID: 37895467 PMCID: PMC10608002 DOI: 10.3390/life13102087] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
An emergence of evidence suggests that severe COVID-19 is associated with an increased risk of developing breast and gastrointestinal cancers. The aim of this research was to assess the risk of heart tumors development in patients who have had COVID-19. METHODS A comparative analysis of 173 heart tumors was conducted between 2016 and 2023. Immunohistochemical examination with antibodies against spike SARS-CoV-2 was performed on 21 heart tumors: 10 myxomas operated before 2020 (the control group), four cardiac myxomas, one proliferating myxoma, three papillary fibroelastomas, two myxofibrosarcomas, one chondrosarcoma resected in 2022-2023. Immunohistochemical analysis with antibodies against CD34 and CD68 was also conducted on the same 11 Post-COVID period heart tumors. Immunofluorescent examination with a cocktail of antibodies against spike SARS-CoV-2/CD34 and spike SARS-CoV-2/CD68 was performed in 2 cases out of 11 (proliferating myxoma and classic myxoma). RESULTS A 1.5-fold increase in the number of heart tumors by 2023 was observed, with a statistically significant increase in the number of myxomas. There was no correlation with vaccination, and no significant differences were found between patients from 2016-2019 and 2021-2023 in terms of gender, age, and cardiac rhythm dis-orders. Morphological examination revealed the expression of spike SARS-CoV-2 in tumor cells, endothelial cells, and macrophages in 10 out of 11 heart tumors. CONCLUSION The detection of SARS-CoV-2 persistence in endothelium and macrophages as well as in tumor cells of benign and malignant cardiac neoplasms, the increase in the number of these tumors, especially cardiac myxomas, after the pandemic by 2023 may indicate a trend toward an increased risk of cardiac neoplasms in COVID-19 patients, which re-quires further research on this issue and a search for new evidence.
Collapse
Affiliation(s)
- Lubov Mitrofanova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Igor Makarov
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Ekaterina Goncharova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Taiana Makarova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Anna Starshinova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| | - Dmitry Kudlay
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 119992 Moscow, Russia;
- Institute of Immunology, 115478 Moscow, Russia
| | - Evgeny Shlaykhto
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (I.M.); (E.G.); (T.M.); (E.S.)
| |
Collapse
|
46
|
Granados AA, Bucher S, Song H, Agrawal A, Chen AT, Peng T, Neff N, Pisco AO, Huang F, Wang B. Single-nuclei characterization of pervasive transcriptional signatures across organs in response to COVID-19. eLife 2023; 12:e81090. [PMID: 37830426 PMCID: PMC10575628 DOI: 10.7554/elife.81090] [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: 06/15/2022] [Accepted: 07/16/2023] [Indexed: 10/14/2023] Open
Abstract
Background Infection by coronavirus SARS-CoV2 is a severe and often deadly disease that has implications for the respiratory system and multiple organs across the human body. While the effects in the lung have been extensively studied, less is known about the impact COVID-19 has across other organs. Methods Here, we contribute a single-nuclei RNA-sequencing atlas comprising six human organs across 20 autopsies where we analyzed the transcriptional changes due to COVID-19 in multiple cell types. The integration of data from multiple organs enabled the identification of systemic transcriptional changes. Results Computational cross-organ analysis for endothelial cells and macrophages identified systemic transcriptional changes in these cell types in COVID-19 samples. In addition, analysis of gene modules showed enrichment of specific signaling pathways across multiple organs in COVID-19 autopsies. Conclusions Altogether, the COVID Tissue Atlas enables the investigation of both cell type-specific and cross-organ transcriptional responses to COVID-19, providing insights into the molecular networks affected by the disease and highlighting novel potential targets for therapies and drug development. Funding The Chan-Zuckerberg Initiative, The Chan-Zuckerberg Biohub.
Collapse
Affiliation(s)
| | - Simon Bucher
- Division of Gastroenterology, Department of Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Hanbing Song
- Department of Medicine, San Francisco Veterans Affairs Medical Center, University of California San FranciscoSan FranciscoUnited States
| | | | | | - Tien Peng
- Yale UniversityNew HavenUnited States
| | - Norma Neff
- Chan-Zuckerberg BiohubSan FranciscoUnited States
| | | | - Franklin Huang
- Department of Medicine, San Francisco Veterans Affairs Medical Center, University of California San FranciscoSan FranciscoUnited States
| | - Bruce Wang
- Division of Gastroenterology, Department of Medicine, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
47
|
Keur N, Saridaki M, Ricaño-Ponce I, Netea MG, Giamarellos-Bourboulis EJ, Kumar V. Analysis of inflammatory protein profiles in the circulation of COVID-19 patients identifies patients with severe disease phenotypes. Respir Med 2023; 217:107331. [PMID: 37364721 PMCID: PMC10290733 DOI: 10.1016/j.rmed.2023.107331] [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: 02/13/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND The coronavirus disease (COVID-19) caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) can present with a broad range of clinical manifestations, ranging from asymptomatic to severe multiple organ failure. The severity of the disease can vary depending on factors such as age, sex, ethnicity, and pre-existing medical conditions. Despite multiple efforts to identify reliable prognostic factors and biomarkers, the predictive capacity of these markers for clinical outcomes remains poor. Circulating proteins, which reflect the active mechanisms in an individual, can be easily measured in clinical practice and therefore may be useful as biomarkers for COVID-19 disease severity. In this study, we sought to identify protein biomarkers and endotypes for COVID-19 severity and evaluate their reproducibility in an independent cohort. METHODS We investigated a cohort of 153 Greek patients with confirmed SARS-CoV-2 infection in which plasma protein levels were measured using the Olink Explore 1536 panel, which consists of 1472 proteins. We compared the protein profiles from severe and moderate COVID-19 patients to identify proteins associated with disease severity. To evaluate the reproducibility of our findings, we compared the protein profiles of 174 patients with comparable COVID-19 severities in a US COVID-19 cohort to identify proteins consistently correlated with COVID-19 severity in both groups. RESULTS We identified 218 differentially regulated proteins associated with severity, 20 proteins were also replicated in an external cohort which we used for validation. Moreover, we performed unsupervised clustering of patients based on 97 proteins with the highest log2 fold changes in order to identify COVID-19 endotypes. Clustering of patients based on differentially regulated proteins revealed the presence of three clinical endotypes. While endotypes 2 and 3 were enriched for severe COVID-19 patients, endotypes 3 represented the most severe form of the disease. CONCLUSIONS These results suggest that identified circulating proteins may be useful for identifying COVID-19 patients with worse outcomes, and this potential utility may extend to other populations. TRIAL REGISTRATION NCT04357366.
Collapse
Affiliation(s)
- Nick Keur
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Maria Saridaki
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Isis Ricaño-Ponce
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania.
| | | | - Vinod Kumar
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Genetics, the Netherlands; Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Deralakatte, Mangalore, India.
| |
Collapse
|
48
|
Tomalka JA, Owings A, Galeas-Pena M, Ziegler CG, Robinson TO, Wichman TG, Laird H, Williams HB, Dhaliwal NS, Everman S, Zafar Y, Shalek AK, Horwitz BH, Ordovas-Montanes J, Glover SC, Gibert Y. Enhanced production of eicosanoids in plasma and activation of DNA damage pathways in PBMCs are correlated with the severity of ancestral COVID-19 infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.14.23295549. [PMID: 37745424 PMCID: PMC10516085 DOI: 10.1101/2023.09.14.23295549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Background Many questions remain unanswered regarding the implication of lipid metabolites in severe SARS-CoV-2 infections. By re-analyzed sequencing data from the nasopharynx of a previously published cohort, we found that alox genes, involved in eicosanoid synthesis, were up-regulated in high WHO score patients, especially in goblet cells. Herein, we aimed to further understand the roles played by eicosanoids during severe SARS-CoV-2 infection. Methods and findings We performed a total fatty acid panel on plasma and bulk RNA-seq analysis on peripheral blood mononuclear cells (PBMCs) collected from 10 infected and 10 uninfected patients. Univariate comparison of lipid metabolites revealed that lipid metabolites were increased in SARS-CoV-2 patients including the lipid mediators Arachidonic Acid (AA) and Eicosapentaenoic Acid (EPA). AA, EPA and the fatty acids Docosahexaenoic acid (DHA) and Docosapentaenoic acid (DPA), were positively correlated to WHO disease severity score. Transcriptomic analysis demonstrated that COVID-19 patients can be segregated based on WHO scores. Ontology, KEGG and Reactome analysis identified pathways enriched for genes related to innate immunity, interactions between lymphoid and nonlymphoid cells, interleukin signaling and, cell cycling pathways. Conclusions Our study offers an association between nasopharynx mucosa eicosanoid genes expression, specific serum inflammatory lipids and, subsequent DNA damage pathways activation in PBMCs to severity of COVID-19 infection.
Collapse
Affiliation(s)
- Jeffrey A. Tomalka
- Dept. of Pathology and Laboratory Medicine. Emory University School of Medicine. Atlanta, GA, USA
| | - Anna Owings
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michelle Galeas-Pena
- Department of Medicine, Section of Gastroenterology and Hepatology, Tulane University School of Medicine. New Orleans, LA, USA
| | - Carly G.K. Ziegler
- Program in Health Sciences & Technology, Harvard Medical School & MIT, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tanya O. Robinson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Thomas G. Wichman
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hannah Laird
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Haley B. Williams
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Neha S. Dhaliwal
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Steven Everman
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yousaf Zafar
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alex K. Shalek
- Program in Health Sciences & Technology, Harvard Medical School & MIT, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Bruce H. Horwitz
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, MA, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA, USA
| | - Jose Ordovas-Montanes
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA, USA
| | - Sarah C. Glover
- Department of Medicine, Section of Gastroenterology and Hepatology, Tulane University School of Medicine. New Orleans, LA, USA
- Dept. of Cell and Molecular Biology; Cancer Center and Research Institute. University of Mississippi Medical Center. Jackson, MS, USA
| | - Yann Gibert
- Dept. of Cell and Molecular Biology; Cancer Center and Research Institute. University of Mississippi Medical Center. Jackson, MS, USA
| |
Collapse
|
49
|
Wan EYF, Zhang R, Mathur S, Yan VKC, Lai FTT, Chui CSL, Li X, Wong CKH, Chan EWY, Lau CS, Wong ICK. Post-acute sequelae of COVID-19 in older persons: multi-organ complications and mortality. J Travel Med 2023; 30:taad082. [PMID: 37310901 DOI: 10.1093/jtm/taad082] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/29/2023] [Accepted: 06/10/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Evidence on long-term associations between coronavirus disease 2019 (COVID-19) and risks of multi-organ complications and mortality in older population is limited. This study evaluates these associations. RESEARCH DESIGN AND METHODS The cohorts included patients aged ≥60 year diagnosed with COVID-19 infection (cases), between 16 March 2020 and 31 May 2021 from the UK Biobank; and between 01 April 2020 and 31 May 2022 from the electronic health records in Hong Kong. Each patient was randomly matched with individuals without COVID-19 infection based on year of birth and sex and were followed for up to 18 months until 31 August 2021 for UKB, and up to 28 months until 15 August 2022 for HK cohort. Patients with COVID-19 infection over 6 months after the date of last dose of vaccination and their corresponding controls were excluded from our study. Characteristics between cohorts were further adjusted with Inverse Probability Treatment Weighting. For evaluating long-term association of COVID-19 with multi-organ disease complications and mortality after 21-days of diagnosis, Cox regression was employed. RESULT 10,759 (UKB) and 165,259 (HK) older adults with COVID-19 infection with matched 291,077 (UKB) and 1,100,394 (HK) non-COVID-19-diagnosed older adults were recruited. Older adults with COVID-19 were associated with a significantly higher risk of cardiovascular outcomes [major cardiovascular disease (stroke, heart failure and coronary heart disease): hazard ratio(UKB): 1.4 (95% Confidence interval: 1.1,1.6), HK:1.2 (95% CI: 1.1,1.3)]; myocardial infarction: HR(UKB): 1.8 (95% CI: 1.3,2.4), HK:1.2 (95% CI: 1.0,1.4)]; respiratory outcomes [interstitial lung disease: HR(UKB: 3.4 (95% CI: 2.5,4.5), HK: 4.0 (95% CI: 1.3,12.8); chronic pulmonary disease: HR(UKB): 1.7 (95% CI: 1.3,2.2), HK:1.6 (95% CI: 1.3,2.1)]; neuropsychiatric outcomes [seizure: HR(UKB): 2.6 (95% CI: 1.7,4.1), HK: 1.6 (95% CI: 1.2,2.1)]; and renal outcomes [acute kidney disease: HR(UKB): 1.4 (95% CI: 1.1,1.6), HK:1.6 (95% CI: 1.3,2.1)]; and all-cause mortality [HR(UKB): 4.9 (95% CI: 4.4,5.4), HK:2.5 (95% CI: 2.5,2.6)]. CONCLUSION COVID-19 is associated with long-term risks of multi-organ complications in older adults (aged ≥ 60). Infected patients in this age-group may benefit from appropriate monitoring of signs/symptoms for developing these complications.
Collapse
Affiliation(s)
- Eric Yuk Fai Wan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ran Zhang
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sukriti Mathur
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vincent Ka Chun Yan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Francisco Tsz Tsun Lai
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
| | - Celine Sze Ling Chui
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xue Li
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Carlos King Ho Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Esther Wai Yin Chan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Department of Pharmacy, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- The University of Hong Kong Shenzhen Institute of Research and Innovation, Shenzhen 518053, China
| | - Chak Sing Lau
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ian Chi Kei Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Science and Technology Park, Hong Kong, China
- Aston Pharmacy School, Aston University, Birmingham B4 7ET, UK
| |
Collapse
|
50
|
Bandyopadhyay S, Rajan MV, Kaur P, Hariprasad G. Identification of potential biomarkers to predict organ morbidity in COVID-19: A repository based proteomics perspective. Biochem Biophys Rep 2023; 35:101493. [PMID: 37304132 PMCID: PMC10235674 DOI: 10.1016/j.bbrep.2023.101493] [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: 02/14/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/13/2023] Open
Abstract
SARS-CoV-2 causes substantial extrapulmonary manifestations in addition to pulmonary disease. Some of the major organs affected are cardiovascular, hematological and thrombotic, renal, neurological, and digestive systems. These types of muti-organ dysfunctions make it difficult and challenging for clinicians to manage and treat COVID-19 patients. The article focuses to identify potential protein biomarkers that can flag various organ systems affected in COVID-19. Publicly reposited high throughput proteomic data from human serum (HS), HEK293T/17 (HEK) and Vero E6 (VE) kidney cell culture were downloaded from ProteomeXchange consortium. The raw data was analyzed in Proteome Discoverer 2.4 to delineate the complete list of proteins in the three studies. These proteins were analyzed in Ingenuity Pathway Analysis (IPA) to associate them to various organ diseases. The shortlisted proteins were analyzed in MetaboAnalyst 5.0 to shortlist potential biomarker proteins. These were then assessed for disease-gene association in DisGeNET and validated by Protein-protein interactome (PPI) and functional enrichment studies (GO_BP, KEGG and Reactome pathways) in STRING. Protein profiling resulted in shortlisting 20 proteins in 7 organ systems. Of these 15 proteins showed at least 1.25-fold changes with a sensitivity and specificity of 70%. Association analysis further shortlisted 10 proteins with a potential association with 4 organ diseases. Validation studies established possible interacting networks and pathways affected, confirmingh the ability of 6 of these proteins to flag 4 different organ systems affected in COVID-19 disease. This study helps to establish a platform to seek protein signatures in different clinical phenotypes of COVID-19. The potential biomarker candidates that can flag organ systems involved are: (a) Vitamin K-dependent protein S and Antithrombin-III for hematological disorders; (b) Voltage-dependent anion-selective channel protein 1 for neurological disorders; (c) Filamin-A for cardiovascular disorder and, (d) Peptidyl-prolyl cis-trans isomerase A and Peptidyl-prolyl cis-trans isomerase FKBP1A for digestive disorders.
Collapse
Affiliation(s)
- Sabyasachi Bandyopadhyay
- Proteomics Sub-facility, Centralized Core Research Facility, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Madhan Vishal Rajan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Gururao Hariprasad
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|