1
|
Siciliani L, Cappa G, Zattera C, Albi G, Mondelli MU, Marzi L. Altered liver hemodynamics in patients with COVID-19: a cross sectional study. J Ultrasound 2025:10.1007/s40477-025-01012-z. [PMID: 40172816 DOI: 10.1007/s40477-025-01012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/16/2025] [Indexed: 04/04/2025] Open
Abstract
AIMS Abnormalities in liver biochemistry are common in COVID-19 patients. Hepatic vein Doppler waveform, typically triphasic, may become biphasic or monophasic in cirrhosis, correlating with liver dysfunction, fibrosis, inflammation, and portal hypertension. This study investigates liver ultrasound (US) features in COVID-19 patients, correlating hepatic vein Doppler waveform and portal vein velocity (PVV) with inflammatory indexes and clinical outcomes. METHODS Fifty-seven patients with SARS-CoV-2 infection participated in a crosssectional study. Bedside upper abdomen US evaluations, including B-mode and Doppler, were conducted using a convex probe. Hepatic vein Doppler waveforms were classified as triphasic, biphasic, or monophasic, and the hepatic vein waveform index (HVWI) was calculated. PVV was measured over three cardiac cycles. Tracings were blindly analyzed by three operators to ensure consistency. RESULTS Low HVWI and high PVV correlated with elevated LDH, ALT, D-dimer, and ferritin (p < 0.05). HVWI showed significant negative correlations with ferritin, D-dimer, and ALT (p < 0.05). D-Dimer and ferritin were higher in patients with biphasic/monophasic waveforms (p < 0.05). High PVV and larger spleen diameters predicted worse respiratory outcomes, including CPAP and tracheal intubation (p < 0.05). Optimal cut-off values for PVV (21.7 cm/s) and spleen diameter (9.84 cm) maximized sensitivity and specificity for predicting these outcomes. FIB-4 scores did not correlate with respiratory outcomes or hepatic hemodynamics (p > 0.05). Hemodynamic alterations were not significantly influenced by the presence of SLD (Steatotic Liver Disease). CONCLUSIONS COVID-19 patients exhibit altered intrahepatic hemodynamics, with hepatic vein waveform abnormalities potentially reflecting liver inflammation and fibrosis. PVV and spleen diameter may serve as non-invasive predictors of respiratory outcomes.
Collapse
Affiliation(s)
- Luisa Siciliani
- Division of Clinical Immunology and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Giovanni Cappa
- Emergency Medicine Unit and Emergency Medicine Postgraduate Training Program, IRCCS Policlinico San Matteo University Hospital, University of Pavia, Pavia, Italy
| | - Caterina Zattera
- Emergency Medicine Unit and Emergency Medicine Postgraduate Training Program, IRCCS Policlinico San Matteo University Hospital, University of Pavia, Pavia, Italy
| | - Giuseppe Albi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Mario Umberto Mondelli
- Division of Clinical Immunology and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Marzi
- Gastroenterology Department, Bolzano Regional Hospital, 39100, Bolzano, Italy
| |
Collapse
|
2
|
Nouanesengsy A, Semesi A, Quach K, Ivanochko D, Byrne W, Hwang M, La Neve MR, Leon-Ponte M, Litosh A, Wisener N, Adeli K, Campigotto A, Grunebaum E, McGeer A, Moraes TJ, Sepiashvili L, Upton J, Julien JP, Allen U. Persistence and decay of neutralizing antibody responses elicited by SARS-CoV-2 infection and hybrid immunity in a Canadian cohort. Microbiol Spectr 2025; 13:e0133324. [PMID: 39969224 PMCID: PMC11960127 DOI: 10.1128/spectrum.01333-24] [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: 06/19/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
A major challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been assessing the intensity, dynamics, and determinants of the antibody responses after infection and/or vaccination. Therefore, we aimed to characterize the longitudinal dynamics of the antibody responses among naturally infected individuals and individuals who achieved hybrid immunity in a large Canadian cohort. We demonstrate that anti-Spike IgGs and neutralizing antibody dynamics vary greatly among individuals with COVID-19, in peak antibody levels, rate of waning, and longevity of the antibody response. Additionally, we found an association between robust antibody responses and individuals with severe COVID-19 clinical symptoms during the first-month post-symptom onset. For individuals who achieved hybrid immunity, a robust increase in anti-S1 IgGs and neutralizing antibodies followed the first vaccination dose; however, there was a minimal increase in the anti-S1 IgGs and neutralizing antibody titers after administration of the second dose of the vaccine. Furthermore, neutralizing antibodies elicited by the wild-type virus alone were largely ineffective against emerging variants of concern in our natural infection-only cohort, in contrast to a much broader and more robust neutralization profile observed in individuals who achieved hybrid immunity. Our findings emphasize the need for global SARS-CoV-2 vaccination efforts to further sustain protective immune responses required to minimize viral spread and disease severity in the population. As SARS-CoV-2 variants continue to emerge, understanding the interplay between previous infections, vaccine durability, and virus evolution will be critical for guiding ongoing vaccination strategies. IMPORTANCE A major challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been assessing the intensity, dynamics, and determinants of the antibody response after infection and/or vaccination. Our paper addresses this in a large Canadian cohort with antibody responses that were generated by natural infection as well as vaccine in some persons studied.
Collapse
Affiliation(s)
- Amy Nouanesengsy
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Anthony Semesi
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Kim Quach
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Walter Byrne
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matthew Hwang
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maria-Rosa La Neve
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matilde Leon-Ponte
- Division of Allergy and Immunology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alice Litosh
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicole Wisener
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Khosrow Adeli
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Aaron Campigotto
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Eyal Grunebaum
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tunenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Theo J. Moraes
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lusia Sepiashvili
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Upton
- Division of Allergy and Immunology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Upton Allen
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Marasco G, Hod K, Colecchia L, Cremon C, Barbaro MR, Cacciari G, Falangone F, Kagramanova A, Bordin D, Drug V, Miftode E, Fusaroli P, Mohamed SY, Ricci C, Bellini M, Rahman MM, Melcarne L, Santos J, Lobo B, Bor S, Yapali S, Akyol D, Sapmaz FP, Urun YY, Eskazan T, Celebi A, Kacmaz H, Ebik B, Binicier HC, Bugdayci MS, Yağcı MB, Pullukcu H, Kaya BY, Tureyen A, Hatemi İ, Koc ES, Sirin G, Calıskan AR, Bengi G, Alıs EE, Lukic S, Trajkovska M, Dumitrascu D, Pietrangelo A, Corradini E, Simren M, Sjolund J, Tornkvist N, Ghoshal UC, Kolokolnikova O, Colecchia A, Serra J, Maconi G, De Giorgio R, Danese S, Portincasa P, Di Sabatino A, Maggio M, Philippou E, Lee YY, Salvi D, Venturi A, Borghi C, Zoli M, Gionchetti P, Viale P, Stanghellini V, Barbara G. Long-Term Impact of COVID-19 on Disorders of Gut-Brain Interaction: Incidence, Symptom Burden, and Psychological Comorbidities. United European Gastroenterol J 2025. [PMID: 40119532 DOI: 10.1002/ueg2.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the potential exacerbation of gastrointestinal symptoms in patients with disorders of gut-brain interaction (DGBIs). However, the distinct symptom trajectories and psychological burden in patients with post-COVID-19 DGBIs compared with patients with pre-existing irritable bowel syndrome (IBS)/functional dyspepsia (FD) and non-DGBI controls remain poorly understood. OBJECTIVES To examine the long-term gastrointestinal symptom progression and psychological comorbidities in patients with post-COVID-19 DGBI, patients with pre-existing IBS/FD and non-DGBI controls. METHODS This post hoc analysis of a prospective multicenter cohort study reviewed patient charts for demographic data and medical history. Participants completed the Gastrointestinal Symptom Rating Scale at four time points: baseline, 1, 6, and 12 months, and the Hospital Anxiety and Depression Scale at 6 and 12 months. The cohort was divided into three groups: (1) post-COVID-19 DGBIs (2) non-DGBI, and (3) pre-existing IBS/FD, with the post-COVID-19 DGBIs group compared to the latter two control groups. RESULTS Among 599 eligible patients, 27 (4.5%) were identified as post-COVID-19 DGBI. This group experienced worsening abdominal pain, hunger pain, heartburn, and acid regurgitation, unlike symptom improvement or stability in non-DGBI controls (p < 0.001 for all symptoms, except hunger pain, p = 0.001). While patients with pre-existing IBS/FD improved in most gastrointestinal symptoms but worsened in constipation and incomplete evacuation, patients with post-COVID-19 DGBI exhibited consistent symptom deterioration across multiple gastrointestinal domains. Anxiety and depression remained unchanged in patients with post-COVID-19 DGBI, contrasting with significant reductions in controls (non-DGBI: p = 0.003 and p = 0.057; pre-existing IBS/FD: p = 0.019 and p = 0.007, respectively). CONCLUSIONS COVID-19 infection is associated with the development of newly diagnosed DGBIs and distinct symptom trajectories when compared with patients with pre-existing IBS/FD. Patients with post-COVID-19 DGBI experience progressive gastrointestinal symptom deterioration and persistent psychological distress, underscoring the need for tailored management strategies for this unique subgroup.
Collapse
Affiliation(s)
- Giovanni Marasco
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Keren Hod
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
- Assuta Medical Centers, Tel Aviv, Israel
| | - Luigi Colecchia
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Cesare Cremon
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Giulia Cacciari
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Falangone
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, University Sapienza, Rome, Italy
| | - Anna Kagramanova
- A. S. Loginov Moscow Clinical Scientific Center, Moscow, Russia
- Research Institute of Health Organization and Medical Management, Moscow, Russia
| | - Dmitry Bordin
- A. S. Loginov Moscow Clinical Scientific Center, Moscow, Russia
- Tver State Medical University, Tver, Russia
- Russian University of Medicine, Moscow, Russia
| | - Vasile Drug
- Department of Gastroenterology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Egidia Miftode
- Department of Infectious Diseases, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | | | - Salem Youssef Mohamed
- Gastroenterology and Hepatology Unit, Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Chiara Ricci
- Department of Experimental and Clinical Sciences, University of Brescia, Brescia, Italy
| | | | - M Masudur Rahman
- Sheikh Russel National Gastroliver Institute and Hospital, Dhaka, Bangladesh
| | - Luigi Melcarne
- Sabadell - CIBEREHD Centro de Investigación Biomédica en Red, Hospital Universitari Parc Taulí, Barcelona, Spain
| | - Javier Santos
- Gastroenterology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Hospital Campus, Barcelona, Spain
- Digestive Physiology and Physiopathology Research Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Hepáticas y Digestivas (CIBERhed), Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Lobo
- Ege University Division of Gastroenterology, Izmir, Turkey
| | - Serhat Bor
- Ege University Division of Gastroenterology, Izmir, Turkey
| | - Suna Yapali
- Division of Gastroenterology, Acibadem University, Altunizade Acibadem Hospital, Istanbul, Turkey
| | - Deniz Akyol
- Ege University Department of Infectious Diseases, Izmir, Turkey
| | - Ferdane Pirincci Sapmaz
- Division of Gastroenterology, University of Health Sciences, Keciören Education and Research Hospital, Keciören, Turkey
| | - Yonca Yilmaz Urun
- Division of Gastroenterology, Eskisehir City Hospital, Eskisehir, Turkey
| | - Tugce Eskazan
- Cerrahpasa Faculty of Medicine, Division of Gastroenterology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Altay Celebi
- Division of Gastroenterology, Kocaeli University, Kocaeli, Turkey
| | - Huseyin Kacmaz
- Division of Gastroenterology, Adiyaman Education and Research Hospital, Adiyaman, Turkey
| | - Berat Ebik
- Division of Gastroenterology, University of Health Sciences, Diyabakır Gazi Yasargil Education and Research Hospital, Diyarbakır, Turkey
| | | | - Mehmet Sait Bugdayci
- Division of Gastroenterology, İstanbul Aydın University Florya Liv Hospital, Istanbul, Turkey
| | | | - Husnu Pullukcu
- Ege University Department of Infectious Diseases, Izmir, Turkey
| | | | - Ali Tureyen
- Division of Gastroenterology, Eskisehir City Hospital, Eskisehir, Turkey
| | - İbrahim Hatemi
- Cerrahpasa Faculty of Medicine, Division of Gastroenterology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Elif Sitre Koc
- Division of Gastroenterology, Acibadem University, Altunizade Acibadem Hospital, Istanbul, Turkey
| | - Goktug Sirin
- Division of Gastroenterology, Kocaeli University, Kocaeli, Turkey
| | - Ali Riza Calıskan
- Division of Gastroenterology, Adiyaman Education and Research Hospital, Adiyaman, Turkey
| | - Goksel Bengi
- Division of Gastroenterology, Dokuz Eylül University, Izmir, Turkey
| | - Esra Ergun Alıs
- Department of Infectious Diseases, İstanbul Aydın University Florya Liv Hospital, Istanbul, Turkey
| | - Snezana Lukic
- Clinic for Gastroenterohepatology, University Clinical Centre of Serbia, Belgrade, Serbia
| | | | - Dan Dumitrascu
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Antonello Pietrangelo
- Internal Medicine Unit, Modena University Hospital, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Corradini
- Internal Medicine Unit, Modena University Hospital, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | - Uday C Ghoshal
- Institute of Gastrosciences and Liver Transplantation, Apollo Multispeciality Hospitals, Kolkata, India
| | | | | | - Jordi Serra
- CIBERehd, University Hospital Germans Trias i Pujol, Barcelona, Spain
| | - Giovanni Maconi
- Gastroenterology Unit, Department of Biomedical and Clinical Sciences, L.Sacco University Hospital, University of Milan, Milan, Italy
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, Milano, Italy
| | - Piero Portincasa
- Division of Internal Medicine "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area, (DiMePre-J) University of Bari "Aldo Moro", Bari, Italy
| | - Antonio Di Sabatino
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Marcello Maggio
- Geriatric Clinic Unit, Medical Geriatric Rehabilitative Department, University Hospital of Parma, Parma, Italy
| | - Elena Philippou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - Yeong Yeh Lee
- School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Daniele Salvi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Claudio Borghi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Marco Zoli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Paolo Gionchetti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Vincenzo Stanghellini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
4
|
Thieulent CJ, Balasuriya UBR, Tseng A, Crossland NA, Stephens JM, Dittmar W, Staszkiewicz J, Richt JA, Carossino M. Diabetes exacerbates SARS-CoV-2 replication through ineffective pulmonary interferon responses, delayed cell-mediated immunity, and disruption of leptin signaling. Front Cell Infect Microbiol 2025; 15:1513687. [PMID: 40125513 PMCID: PMC11925909 DOI: 10.3389/fcimb.2025.1513687] [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: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 03/25/2025] Open
Abstract
Comorbidities, including obesity and type 2 diabetes mellitus (T2DM), are associated with increased disease severity and mortality following SARS-CoV-2 infection. Here, we investigated virus-host interactions under the effects of these comorbidities in diet-induced obesity (DIO) and leptin receptor-deficient (T2DM) mice following infection with SARS-CoV-2. DIO mice, as well as their lean counterparts, showed limited susceptibility to SARS-CoV-2 infection. In contrast, T2DM mice showed exacerbated pulmonary SARS-CoV-2 replication and delayed viral clearance associated with down-regulation of innate and adaptative immune gene signatures, ineffective type I interferon response, and delayed SARS-CoV-2-specific cell-mediated immune responses. While T2DM mice showed higher and prolonged SARS-CoV-2-specific immunoglobulin isotype responses compared to their lean counterparts, neutralizing antibody levels were equivalent. By silencing the leptin receptor in vitro using a human alveolar epithelial cell line, we observed an increase in SARS-CoV-2 replication and type I interferons. Altogether, our data provides for the first time evidence that disruption of leptin receptor signaling leading to obesity and T2DM induces altered type I interferon and cell-mediated responses against SARS-CoV-2, mediating increased viral replication and delayed clearance. These data shed light on the alteration of the innate immune pathway in the lung using in-depth transcriptomic analysis and on adaptive immune responses to SARS-CoV-2 under T2DM conditions. Finally, this study provides further insight into this risk factor aggravating SARS-CoV-2 infection and understanding the underlying cellular mechanisms that could help identify potential intervention points for this at-risk population.
Collapse
MESH Headings
- Animals
- COVID-19/immunology
- COVID-19/virology
- COVID-19/metabolism
- Mice
- Virus Replication
- Receptors, Leptin/metabolism
- Receptors, Leptin/genetics
- SARS-CoV-2/immunology
- Signal Transduction
- Humans
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/metabolism
- Obesity/immunology
- Leptin/metabolism
- Lung/immunology
- Lung/virology
- Immunity, Cellular
- Interferon Type I/metabolism
- Antibodies, Neutralizing/immunology
- Disease Models, Animal
- Mice, Inbred C57BL
- Male
- Immunity, Innate
- Mice, Knockout
Collapse
Affiliation(s)
- Côme J. Thieulent
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Udeni B. R. Balasuriya
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Anna Tseng
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Nicholas A. Crossland
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Jacqueline M. Stephens
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Wellesley Dittmar
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Jaroslaw Staszkiewicz
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Juergen A. Richt
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
5
|
Pan D, Diaz JL, Weidman K, Graham J, Goyal P, Rajan M, Lau J, Pinheiro L, Rachid L, Simmons W, Schenck EJ, Safford MM, Lief L. Social Networks as a Key Health Determinant in Acute Illness Recovery: A Lesson from the COVID-19 Pandemic. Am J Med 2025; 138:541-549.e5. [PMID: 38677397 PMCID: PMC11502506 DOI: 10.1016/j.amjmed.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND The COVID-19 pandemic highlighted the importance of considering social determinants of health in health outcomes. Within this spectrum of determinants, social networks garnered attention as the pandemic highlighted the negative effects of social isolation in the context of social distancing measures. Postpandemic, examining the role social networks play in COVID-19 recovery can help guide patient care and shape future health policies. This study aimed to investigate the relationship between social networks and self-rated health change, as well as physical function, in patients recovering from COVID-19 pneumonia. METHODS This was a retrospective cohort study utilizing clinical data from 2 New York City hospitals and a 9-month follow-up survey of COVID-19 pneumonia survivors. We evaluated a composite Social Network Score from the 6-item Lubben Social Network Scale and its association with 2 outcomes: 1) self-rated health change and 2) physical function. RESULTS A total of 208 patients were included in this study. A 1-point increase in the Social Network Score was associated with greater odds of both same or improved self-rated health change (odds ratio [OR] 1.07, 95% CI 1.02-1.12, P = .01), as well as unimpaired physical function (OR 1.08, 95% CI 1.03-1.14, P < .01). CONCLUSION This study emphasized the importance of social networks as a social determinant of health among patients recovering from COVID-19 hospitalization. Targeted interventions to enhance social networks may benefit not only COVID-19 patients but also individuals recovering from other acute illnesses.
Collapse
Affiliation(s)
- Di Pan
- Division of Pulmonary and Critical Care, Department of Medicine, Weill Cornell Medical College, New York, N.Y..
| | - Jihui L Diaz
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, Penn
| | - Karissa Weidman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, N.Y
| | - Julia Graham
- Division of Pulmonary and Critical Care, Department of Medicine, Weill Cornell Medical College, New York, N.Y
| | - Parag Goyal
- Division of Cardiology, Department of Medicine, Weill Cornell Medical College, New York, N.Y
| | - Mangala Rajan
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medical College, New York, N.Y
| | - Jennifer Lau
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medical College, New York, N.Y
| | - Laura Pinheiro
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medical College, New York, N.Y
| | - Leena Rachid
- Stritch School of Medicine at Loyola University Chicago, Maywood, Ill
| | - Will Simmons
- Department of Population Health Sciences, Weill Cornell Medical College, New York, N.Y
| | - Edward J Schenck
- Division of Pulmonary and Critical Care, Department of Medicine, Weill Cornell Medical College, New York, N.Y
| | - Monika M Safford
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medical College, New York, N.Y
| | - Lindsay Lief
- Division of Pulmonary and Critical Care, Department of Medicine, Weill Cornell Medical College, New York, N.Y
| |
Collapse
|
6
|
Pérez-Gisbert L, Morales-García C, Sánchez-Martínez JA, González-Gutiérrez MV, Valenza MC, Torres-Sánchez I. Severity Matters: How COVID-19 Severity Impacts Long-Term Effects on Symptoms, Physical Activity and Functionality-An Observational Study. Healthcare (Basel) 2025; 13:333. [PMID: 39942522 PMCID: PMC11817242 DOI: 10.3390/healthcare13030333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES The existing literature has described the common symptoms and long-term effects of coronavirus disease (COVID-19). However, there is a lack of detailed information on how different degrees of disease severity affect survivors differently. This study aims to fill that gap by evaluating the symptoms, physical activity, and functionality of COVID-19 survivors across a spectrum of severity levels, comparing them with those of healthy individuals. METHODS An observational study was carried out following the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) criteria and checklist. Participants were divided into 5 groups based on COVID-19 severity according to the World Health Organization classification: healthy (COVID-19-negative), mild (symptomatic without pneumonia or dyspnoea), moderate (pneumonia and dyspnoea without hospitalisation), severe (severe pneumonia requiring hospitalisation), and critical (severe pneumonia with admission to the intensive care unit). Descriptive variables, symptoms (Fatigue Borg Scale, Fatigue Impact Scale, Fatigue Severity Scale, Dyspnoea Borg Scale, Visual Analogue Scale, Hospital Anxiety and Depression Scale, and European Quality of Life-5 Dimensions), physical activity (the International Physical Activity Questionnaire) and functionality (Patient-Specific Functional Scale, Short Physical Performance Battery, Arm Curl test, and 2 min step test) were measured. RESULTS A total of 304 participants were included: healthy (n = 42), mild (n = 143), moderate (n = 49), severe (n = 52), and critical (n = 18) COVID-19 patients. The impact of COVID-19 on surviving patients varies significantly with the severity of the disease. The results show that the hospitalisation time, age, and comorbidities of the patients are greater in those with a greater severity of the disease. Patients with more severe COVID-19 also experience greater frailty, dysphagia, fatigue, dyspnoea, and pain. Additionally, those with severe cases have poorer overall health, reduced physical activity, and diminished functionality. No evidence of post-COVID-19 anxiety or depression is found in the sample, even considering the timeframe between the negative test and the assessment. CONCLUSIONS Patients with higher COVID-19 severity (severe or critical) experience more symptoms than those with lower COVID-19 severity (mild or moderate). Additionally, those with severe cases have poorer overall health, reduced physical activity and diminished functionality. Register: Clinicaltrials.gov: NCT05731817.
Collapse
Affiliation(s)
- Laura Pérez-Gisbert
- Physical Therapy Department, Faculty of Health Sciences, University of Granada, Avenida de la Ilustración nº 60, 18016 Granada, Spain; (L.P.-G.); (M.C.V.)
| | - Concepción Morales-García
- Pneumology Service, Virgen de las Nieves University Hospital, Avenida de las Fuerzas Armadas nº 2, 18014 Granada, Spain; (C.M.-G.); (J.A.S.-M.); (M.V.G.-G.)
| | - José Antonio Sánchez-Martínez
- Pneumology Service, Virgen de las Nieves University Hospital, Avenida de las Fuerzas Armadas nº 2, 18014 Granada, Spain; (C.M.-G.); (J.A.S.-M.); (M.V.G.-G.)
| | - María Victoria González-Gutiérrez
- Pneumology Service, Virgen de las Nieves University Hospital, Avenida de las Fuerzas Armadas nº 2, 18014 Granada, Spain; (C.M.-G.); (J.A.S.-M.); (M.V.G.-G.)
| | - Marie Carmen Valenza
- Physical Therapy Department, Faculty of Health Sciences, University of Granada, Avenida de la Ilustración nº 60, 18016 Granada, Spain; (L.P.-G.); (M.C.V.)
| | - Irene Torres-Sánchez
- Physical Therapy Department, Faculty of Health Sciences, University of Granada, Avenida de la Ilustración nº 60, 18016 Granada, Spain; (L.P.-G.); (M.C.V.)
| |
Collapse
|
7
|
De Souza Dos Santos G, Alves de Carvalho França de Macedo V, Oliniski Reikdal S, Graf ME, Mario Martin B, Joaquim Meier M. Ventilator-associated pneumonia risk factors in patients with severe COVID-19 in southern Brazil: A retrospective observational study. Infect Dis Health 2025; 30:38-49. [PMID: 39168742 DOI: 10.1016/j.idh.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
BACKGOUND During the SARS-CoV-2 pandemic, a significant number of critical patients required ventilatory assistance in health institutions. In this context, Ventilator-Associated Pneumonia (VAP) was the most prevalent nosocomial infection among critically ill patients. We aimed to analyze the occurrence of VAP in critically ill patients with SARS-CoV-2 and the risk factors associated with the outcome. METHOD This is a multicenter, retrospective cohort study which included patients ≥18 years old, diagnosed with COVID-19, admitted to intensive care units (ICU) and who received invasive mechanical ventilation (MV) for >2 consecutive days. The associations between the variables were initially tested, and those that showed potential associations (p<0.05) were included in the multivariate logistic regression model. RESULTS One third of patients had an episode of VAP, with an incidence density of 34.97 cases per 1000 MV days. In addition, 42.37% (50) of the microorganisms causing VAP were multidrug-resistant, predominantly gram-negative bacteria (61.32%). More than 50% of participants developed healthcare-associated infections and 243 (73.64%) died. The factors associated with greater chances of VAP were: prone position (OR= 3.77), BMI 25-29.9 kg/m2 (OR= 4.76), pressure injury (OR= 4.41), length of stay in the ICU (OR= 1.06), positive tracheal aspirate before VAP (OR= 5.41) and dyspnea (OR= 3.80). CONCLUSIONS Patients with COVID-19 are at high risk of VAP, which leads to an increased risk of death (OR = 2.18). Multiple factors increase the chances of VAP in this population, namely: work overload in health institutions, prone position, prolonged ICU time, infusion of multiple drugs, invasive devices, and in particular, immobility in bed.
Collapse
Affiliation(s)
- Gabriela De Souza Dos Santos
- Specialized Care Planning Board, State Health Department of Parana, Curitiba, 828230-140, Brazil; Postgraduate Nursing Department, Federal University of Parana, Curitiba, 80210-170, Brazil.
| | - Viviane Alves de Carvalho França de Macedo
- Hospital Infection Control and Epidemiology Center, Santa Casa de Curitiba, Curitiba, 80010-030, Brazil; Medicine School Department, Positive University, Curitiba, 81280-330, Brazil; Department of Infectious Diseases, Faculty of Medicine at the University of São Paulo, 01246903, Brazil
| | - Samantha Oliniski Reikdal
- Nursing Department, Dom Bosco Universitary Center, Curitiba, 81010-000, Brazil; Nursing Department, Santa Cruz Universitary Center, Curitiba, 81050-180, Brazil
| | - Maria Esther Graf
- Infection Control Program, Hospital de Clınicas, Curitiba, 80060-900, Brazil; Hospital Infection Control and Epidemiology Center, Trabalhador Hospital, Curitiba, 81050-000, Brazil
| | - Beatris Mario Martin
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland Herston, Brisbane, Queensland 4029, Australia
| | - Marineli Joaquim Meier
- Postgraduate Nursing Department, Federal University of Parana, Curitiba, 80210-170, Brazil
| |
Collapse
|
8
|
Han C, Jung YJ, Park JE, Chung WY, Yoon D. Artificial Intelligence-Based Early Prediction of Acute Respiratory Failure in the Emergency Department Using Biosignal and Clinical Data. Yonsei Med J 2025; 66:121-130. [PMID: 39894045 PMCID: PMC11790410 DOI: 10.3349/ymj.2024.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 02/04/2025] Open
Abstract
PURPOSE Early identification of patients at risk for acute respiratory failure (ARF) could help clinicians devise preventive strategies. Analyzing biosignals with artificial intelligence (AI) can uncover hidden information and variability within time series. We aimed to develop and validate AI models to predict ARF within 72 h after emergency department admission, primarily using high-resolution biosignals collected within 4 h of arrival. MATERIALS AND METHODS Our AI model, built on convolutional recurrent neural networks, combines biosignal feature extraction and sequence modeling. The model was developed and internally validated with data from 5284 admissions [1085 (20.5%) positive for ARF], and externally validated using data from 144 admissions [7 (4.9%) positive for ARF] from another institution. We defined ARF as the application of advanced respiratory support devices. RESULTS Our AI model performed well in predicting ARF, achieving area under the receiver operating characteristic curve (AUROC) of 0.840 and 0.743 in internal and external validations, respectively. It outperformed the Modified Early Warning Score (MEWS) and XGBoost models built only with clinical variables. High predictive ability for mortality was observed, with AUROC up to 0.809. A 10% increase in AI prediction scores was associated with 1.44-fold and 1.42-fold increases in ARF risk and mortality risk, respectively, even after adjusting for MEWS and demographic variables. CONCLUSION Our AI model demonstrates high predictive accuracy and significant associations with clinical outcomes. Our AI model has the potential to promptly aid in triage decisions. Our study shows that using AI to analyze biosignals advances disease detection and prediction.
Collapse
Affiliation(s)
- Changho Han
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea
| | - Yun Jung Jung
- Department of Pulmonology and Critical Care Medicine, Ajou University School of Medicine, Suwon, Korea
| | - Ji Eun Park
- Department of Pulmonology and Critical Care Medicine, Ajou University School of Medicine, Suwon, Korea
| | - Wou Young Chung
- Department of Pulmonology and Critical Care Medicine, Ajou University School of Medicine, Suwon, Korea.
| | - Dukyong Yoon
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea
- Institute for Innovation in Digital Healthcare (IIDH), Severance Hospital, Seoul, Korea
- Center for Digital Health, Yongin Severance Hospital, Yonsei University Health System, Yongin, Korea.
| |
Collapse
|
9
|
Mohammed Ismail W, Fernandez JA, Binder M, Lasho TL, Kim M, Geyer SM, Mazzone A, Finke CM, Mangaonkar AA, Lee JH, Wang L, Kim KH, Simon VA, Rakhshan Rohakthar F, Munankarmy A, Byeon SK, Schwager SM, Harrington JJ, Snyder MR, Robertson KD, Pandey A, Wieben ED, Chia N, Gaspar-Maia A, Patnaik MM. Single-cell multiomics reveal divergent effects of DNMT3A- and TET2-mutant clonal hematopoiesis in inflammatory response. Blood Adv 2025; 9:402-416. [PMID: 39631069 PMCID: PMC11787483 DOI: 10.1182/bloodadvances.2024014467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/28/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
ABSTRACT DNMT3A and TET2 are epigenetic regulator genes commonly mutated in age-related clonal hematopoiesis (CH). Despite having opposed epigenetic functions, these mutations are associated with increased all-cause mortality and a low risk for progression to hematologic neoplasms. Although individual impacts on the epigenome have been described using different model systems, the phenotypic complexity in humans remains to be elucidated. Here, we make use of a natural inflammatory response occurring during coronavirus disease 2019 (COVID-19), to understand the association of these mutations with inflammatory morbidity (acute respiratory distress syndrome [ARDS]) and mortality. We demonstrate the age-independent, negative impact of DNMT3A mutant (DNMT3Amt) CH on COVID-19-related ARDS and mortality. Using single-cell proteogenomics we show that DNMT3A mutations involve myeloid and lymphoid lineage cells. Using single-cell multiomics sequencing, we identify cell-specific gene expression changes associated with DNMT3A mutations, along with significant epigenomic deregulation affecting enhancer accessibility, resulting in overexpression of interleukin-32 (IL-32), a proinflammatory cytokine that can result in inflammasome activation in monocytes and macrophages. Finally, we show with single-cell resolution that the loss of function of DNMT3A is directly associated with increased chromatin accessibility in mutant cells. Hence, we demonstrate the negative prognostic impact of DNMT3Amt CH on COVID-19-related ARDS and mortality. DNMT3Amt CH in the context of COVID-19, was associated with inflammatory transcriptional priming, resulting in overexpression of IL32. This overexpression was secondary to increased chromatic accessibility, specific to DNMT3Amt CH cells. DNMT3Amt CH can thus serve as a potential biomarker for adverse outcomes in COVID-19.
Collapse
Affiliation(s)
- Wazim Mohammed Ismail
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Jenna A. Fernandez
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Moritz Binder
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Terra L. Lasho
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Minsuk Kim
- Department of Laboratory Medicine and Pathology, Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Susan M. Geyer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Amelia Mazzone
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Christy M. Finke
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | | | - Jeong-Heon Lee
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Liguo Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Kwan Hyun Kim
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | - Amik Munankarmy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Susan M. Schwager
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Jonathan J. Harrington
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Melissa R. Snyder
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Keith D. Robertson
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Eric D. Wieben
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Nicholas Chia
- Department of Laboratory Medicine and Pathology, Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Alexandre Gaspar-Maia
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Mrinal M. Patnaik
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
10
|
Kern M, Hamm SR, Pedersen CR, Møller DL, Loft JA, Hasselbalch RB, Heftdal LD, Pries-Heje MM, Perch M, Sørensen SS, Rasmussen A, Garred P, Iversen KK, Bundgaard H, Sabin CA, Nielsen SD. Leukocyte Count in Solid Organ Transplant Recipients After SARS-CoV-2 mRNA Vaccination and Infection. Vaccines (Basel) 2025; 13:103. [PMID: 40006650 PMCID: PMC11860179 DOI: 10.3390/vaccines13020103] [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: 10/30/2024] [Revised: 12/20/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Solid organ transplant (SOT) recipients are at risk of severe COVID-19. Vaccination is an important preventive measure but may have side effects, including decreased leukocyte counts. We aimed to describe the prevalence and relative incidence of decreased leukocyte counts and changes in leukocyte counts before and after SARS-CoV-2 mRNA vaccination and SARS-CoV-2 infection in SOT recipients. METHODS Changes in leukocyte counts from before to after each vaccine dose were investigated using linear mixed models. We determined the prevalence of decreased leukocyte counts before and after each vaccine dose and before and after SARS-CoV-2 infection. Self-controlled case series analysis was used to investigate whether the period after either vaccination or infection was associated with risk of decreased leukocyte count. RESULTS We included 228 adult kidney, lung, and liver transplant recipients. Prior to the first vaccine dose, the mean leukocyte count was 7.3 × 109 cells/L (95% CI 6.9-7.6). Both the leukocyte counts, and the prevalence of decreased leukocyte counts remained unchanged from before to after vaccination regardless of the number of vaccine doses provided. There was no association between vaccination and decreased leukocyte counts (incidence rate ratio (IRR): 0.6; 95% CI: 0.2-2.1; p = 0.461). In contrast, SARS-CoV-2 infection was associated with increased risk of a decreased leukocyte count (IRR: 7.1; 95% CI: 2.8-18.1; p < 0.001). CONCLUSIONS SARS-CoV-2 mRNA vaccination was not associated with risk of decreased leukocyte count and did not affect the prevalence of decreased leukocyte counts in SOT recipients. In contrast, SARS-CoV-2 infection was associated with a higher risk of a decreased leukocyte count.
Collapse
Affiliation(s)
- Marita Kern
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark (S.R.H.)
| | - Sebastian Rask Hamm
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark (S.R.H.)
| | - Christian Ross Pedersen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Dina Leth Møller
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark (S.R.H.)
| | - Josefine Amalie Loft
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark (S.R.H.)
| | - Rasmus Bo Hasselbalch
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Line Dam Heftdal
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark (S.R.H.)
| | - Mia Marie Pries-Heje
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Michael Perch
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Søren Schwartz Sørensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Nephrology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Allan Rasmussen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Peter Garred
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Laboratory of Molecular medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Kasper Karmark Iversen
- Department of Cardiology, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Caroline A. Sabin
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, UCL, Royal Free Campus, Rowland Hill St., London NW3 2PF, UK
| | - Susanne Dam Nielsen
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark (S.R.H.)
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
11
|
Zheng J, Dhakal H, Qing E, Shrestha R, Geller AE, Morrissey SM, Saxena D, Hu X, Li H, Li H, Wilhelmsen K, Wendt LH, Klumpp K, Hume PS, Janssen WJ, Brody R, Palmer KE, Uriarte SM, Ten Eyck P, Meyerholz DK, Merchant ML, McLeish K, Gallagher T, Huang J, Yan J, Perlman S. CXCL12 ameliorates neutrophilia and disease severity in SARS-CoV-2 infection. J Clin Invest 2025; 135:e188222. [PMID: 39773555 PMCID: PMC11827850 DOI: 10.1172/jci188222] [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: 10/18/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025] Open
Abstract
Neutrophils, particularly low-density neutrophils (LDNs), are believed to contribute to acute COVID-19 severity. Here, we showed that neutrophilia can be detected acutely and even months after SARS-CoV-2 infection in patients and mice, while neutrophil depletion reduced disease severity in mice. A key factor in neutrophilia and severe disease in infected mice was traced to the chemokine CXCL12 secreted by bone marrow cells and unexpectedly, endothelial cells. CXCL12 levels were negatively correlated with LDN numbers in longitudinal analyses of patient blood samples. CXCL12 blockade in SARS-CoV-2-infected mice increased blood/lung neutrophil numbers, thereby accelerating disease progression without changing lung virus titers. The exaggerated mortality caused by CXCL12 blockade could be reversed by neutrophil depletion. In addition, blocking interactions between SARS-CoV-2 and angiotensin-converting enzyme 2 (ACE2) reduced CXCL12 levels, suggesting a signal transduction from virus-mediated ACE2 ligation to increased CXCL12 secretion. Collectively, these results demonstrate a previously unappreciated role of CXCL12 in diminishing neutrophilia, including low-density neutrophilia, and its deleterious effects in SARS-CoV-2 infections. The results also support the involvement of SARS-CoV-2-endothelial cell interactions in viral pathogenesis.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Microbiology and Immunology and
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Hima Dhakal
- Department of Microbiology and Immunology and
| | - Enya Qing
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Rejeena Shrestha
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Anne E. Geller
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Samantha M. Morrissey
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Divyasha Saxena
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Xiaoling Hu
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Hong Li
- Functional Immunomics Core, Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Haiyan Li
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | | | - Linder H. Wendt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, USA
| | | | - Patrick S. Hume
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - William J. Janssen
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Rachel Brody
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kenneth E. Palmer
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology and
| | - Silvia M. Uriarte
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Patrick Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, USA
| | | | | | - Kenneth McLeish
- Department of Medicine, Division of Nephrology and Hypertension and
| | - Tom Gallagher
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Jiapeng Huang
- Department of Anesthesiology and Perioperative Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jun Yan
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
12
|
Rizwan M, Cheng K, Gang Y, Hou Y, Wang C. Immunomodulatory Effects of Vitamin D and Zinc on Viral Infection. Biol Trace Elem Res 2025; 203:1-17. [PMID: 38451442 DOI: 10.1007/s12011-024-04139-y] [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: 01/24/2024] [Accepted: 03/02/2024] [Indexed: 03/08/2024]
Abstract
Several nutrients are crucial in enhancing the immune system and preserving the structural integrity of bodily tissue barriers. Vitamin D (VD) and zinc (Zn) have received considerable interest due to their immunomodulatory properties and ability to enhance the body's immune defenses. Due to their antiviral, anti-inflammatory, antioxidative, and immunomodulatory properties, the two nutritional powerhouses VD and Zn are crucial for innate and adaptive immunity. As observed with COVID-19, deficiencies in these micronutrients impair immune responses, increasing susceptibility to viral infections and severe disease. Ensuring an adequate intake of VD and Zn emerges as a promising strategy for fortifying the immune system. Ongoing clinical trials are actively investigating their potential therapeutic advantages. Beyond the immediate context of the pandemic, these micronutrients offer valuable tools for enhancing immunity and overall well-being, especially in the face of future viral threats. This analysis emphasizes the enduring significance of VD and Zn as both treatment and preventive measures against potential viral challenges beyond the current health crisis. The overview delves into the immunomodulatory potential of VD and Zn in combating viral infections, with particular attention to their effects on animals. It provides a comprehensive summary of current research findings regarding their individual and synergistic impacts on immune function, underlining their potential in treating and preventing viral infections. Overall, this overview underscores the need for further research to understand how VD and Zn can modulate the immune response in combatting viral diseases in animals.
Collapse
Affiliation(s)
- Muhammad Rizwan
- College of Fisheries, Huazhong Agriculture University, Wuhan, 430070, China
| | - Ke Cheng
- College of Fisheries, Huazhong Agriculture University, Wuhan, 430070, China
| | - Yang Gang
- College of Fisheries, Huazhong Agriculture University, Wuhan, 430070, China
| | - Yuntao Hou
- College of Fisheries, Huazhong Agriculture University, Wuhan, 430070, China
| | - Chunfang Wang
- College of Fisheries, Huazhong Agriculture University, Wuhan, 430070, China.
| |
Collapse
|
13
|
Adilović M, Hromić-Jahjefendić A, Mahmutović L, Šutković J, Rubio-Casillas A, Redwan EM, Uversky VN. Intrinsic Factors Behind the Long-COVID: V. Immunometabolic Disorders. J Cell Biochem 2025; 126:e30683. [PMID: 39639607 DOI: 10.1002/jcb.30683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/02/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
The complex link between COVID-19 and immunometabolic diseases demonstrates the important interaction between metabolic dysfunction and immunological response during viral infections. Severe COVID-19, defined by a hyperinflammatory state, is greatly impacted by underlying chronic illnesses aggravating the cytokine storm caused by increased levels of Pro-inflammatory cytokines. Metabolic reprogramming, including increased glycolysis and altered mitochondrial function, promotes viral replication and stimulates inflammatory cytokine production, contributing to illness severity. Mitochondrial metabolism abnormalities, strongly linked to various systemic illnesses, worsen metabolic dysfunction during and after the pandemic, increasing cardiovascular consequences. Long COVID-19, defined by chronic inflammation and immune dysregulation, poses continuous problems, highlighting the need for comprehensive therapy solutions that address both immunological and metabolic aspects. Understanding these relationships shows promise for effectively managing COVID-19 and its long-term repercussions, which is the focus of this review paper.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jasmin Šutković
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Mexico
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
14
|
Patrascu R, Dumitru CS, Laza R, Besliu RS, Gug M, Zara F, Laitin SMD. The Role of Age and Comorbidity Interactions in COVID-19 Mortality: Insights from Cardiac and Pulmonary Conditions. J Clin Med 2024; 13:7510. [PMID: 39768431 PMCID: PMC11677844 DOI: 10.3390/jcm13247510] [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: 11/14/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Understanding the interactions between age and comorbidities is crucial for assessing COVID-19 mortality, particularly in patients with cardiac and pulmonary conditions. This study investigates the relationship between comorbidities and mortality outcomes in a cohort of hospitalized COVID-19 patients, emphasizing the interplay of age, cardiac, and pulmonary conditions. Methods: We analyzed a cohort of 3005 patients hospitalized with COVID-19 between 2020 and 2022. Key variables included age, comorbidities (diabetes, cardiac, pulmonary, and neoplasms), and clinical outcomes. Chi-square tests and logistic regression models were used to assess the association between comorbidities and mortality. Stratified analyses by age, diabetes, and pulmonary conditions were conducted to explore interaction effects. Additionally, interaction terms were included in multivariable logistic regression models to evaluate the combined impact of age, comorbidities, and mortality. Results: Cardiac conditions such as hypertension, ischemic cardiopathy, and myocardial infarction showed significant protective effects against mortality in younger patients and in those without pulmonary conditions (p < 0.001). However, these protective effects were diminished in older patients and those with pulmonary comorbidities. Age was found to be a significant modifier of the relationship between cardiac conditions and mortality, with a stronger protective effect observed in patients under the median age (p < 0.001). Pulmonary comorbidities significantly increased the risk of mortality, particularly when co-occurring with cardiac conditions (p < 0.001). Diabetes did not significantly modify the relationship between cardiac conditions and mortality. Conclusions: The findings highlight the complex interactions between age, cardiac conditions, and pulmonary conditions in predicting COVID-19 mortality. Younger patients with cardiac comorbidities show a protective effect against mortality, while pulmonary conditions increase mortality risk, especially in older patients. These insights suggest that individualized risk assessments incorporating age and comorbidities are essential for managing COVID-19 outcomes.
Collapse
Affiliation(s)
- Raul Patrascu
- Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Cristina Stefania Dumitru
- Department of Microscopic Morphology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Ruxandra Laza
- Infectious Diseases University Clinic, Department XIII, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania;
- Clinical Hospital of Infectious Diseases and Pneumology “Dr. Victor Babes”, 300310 Timisoara, Romania;
| | - Razvan Sebastian Besliu
- Epidemiology Clinic, ‘Pius Brinzeu’ Emergency Clinical County Hospital Timisoara, Liviu Rebreanu Boulevard No. 156, 300723 Timisoara, Romania;
| | - Miruna Gug
- Discipline of Genetics, Department of Microscopic Morphology, Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Flavia Zara
- Department of Microscopic Morphology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| | - Sorina Maria Denisa Laitin
- Clinical Hospital of Infectious Diseases and Pneumology “Dr. Victor Babes”, 300310 Timisoara, Romania;
- Epidemiology University Clinic, Department XIII, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| |
Collapse
|
15
|
Rabiei S, Kamali Z, Jamilian P, Jamilian P. Beneficial effects of probiotics to flatten the curve of COVID-19 pandemic: A review. CLINICAL NUTRITION OPEN SCIENCE 2024; 58:348-360. [DOI: 10.1016/j.nutos.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
|
16
|
Khayat O, Basheer M, Derawy M, Assy N. Early Elevated Inflammatory Markers in SARS-CoV-2 Vaccinated Patients Are Associated with Reduced Mortality, Morbidity, and Lung Injury. Life (Basel) 2024; 14:1415. [PMID: 39598212 PMCID: PMC11595982 DOI: 10.3390/life14111415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/08/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Background The development of vaccines against SARS-CoV-2 has proved to be a highly successful strategy. In this work, the aim is to study the effects of the SARS-CoV-2 vaccine on the production of inflammatory markers and how this affect morbidity and mortality. Electronic medical record (EMR) data from 210 patients diagnosed with COVID-19 from November 2020 to June 2021 were collected. The admitted patients were divided into three groups, the one-dose vaccinated, two-dose vaccinated, and the non-vaccinated. All patients were moderate or severe in disease level as defined by the WHO classification. The results show that CRP was 101 ± 5.3, 97 ± 10.8, and 145 ± 17.3 (p < 0.05), fibrinogen 529 ± 16.3, 397 ± 33.8, and 610 ± 15 (p < 0.05), D-dimer 1244 ± 89, 1279 ± 297, and 1615 ± 224 (p < 0.05), ferritin was 1170 ± 122, 999 ± 202, and 1663 ± 409 (p < 0.05), IL-6 was 196 ± 12, 96 ± 5, and 580 ± 402 (NS), for the non-vaccinated, one-dose vaccinated, and two-dose vaccinated groups, respectively. The high level of CRP up to 150-200 mg/dL was more common among the surviving vaccinated patients. Oxygen supplementation, mechanical ventilation, and mortality were higher in the non-vaccinated group. Blood urea nitrogen (BUN) level was higher in the vaccinated patients, 25 ± 0.14 vs. 33 ± 6.15, respectively (p < 0.05). Inflammation markers were significantly higher in the vaccinated groups compared to non-vaccinated groups. On the other hand, extremely high levels of CRP (>200 mg/dL) were correlated with high mortality incidence.
Collapse
Affiliation(s)
- Osama Khayat
- Internal Medicine Department, Galilee Medical Center, Nahariya 2210001, Israel; (M.D.); (N.A.)
| | - Maamoun Basheer
- Internal Medicine Department, Galilee Medical Center, Nahariya 2210001, Israel; (M.D.); (N.A.)
- Azrieli Bar-Ilan Faculty of Medicine, Safad 1311502, Israel
| | - Mayss Derawy
- Internal Medicine Department, Galilee Medical Center, Nahariya 2210001, Israel; (M.D.); (N.A.)
| | - Nimer Assy
- Internal Medicine Department, Galilee Medical Center, Nahariya 2210001, Israel; (M.D.); (N.A.)
- Azrieli Bar-Ilan Faculty of Medicine, Safad 1311502, Israel
| |
Collapse
|
17
|
Norton TD, Thakur M, Ganguly S, Ali S, Chao J, Waldron A, Xiao J, Patel Y, Turner KC, Davis JD, Irvin SC, Pan C, Atmodjo-Watkins D, Hooper AT, Hamilton JD, Subramaniam D, Bocchini JA, Kowal B, DiCioccio AT, Bhore R, Geba GP, Cox E, Braunstein N, Dakin P, Herman GA. Assessing the safety and pharmacokinetics of casirivimab and imdevimab (CAS+IMD) in a cohort of pregnant outpatients with COVID-19: results from an adaptive, multicentre, randomised, double-blind, phase 1/2/3 study. BMJ Open 2024; 14:e087431. [PMID: 39384241 PMCID: PMC11474922 DOI: 10.1136/bmjopen-2024-087431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/19/2024] [Indexed: 10/11/2024] Open
Abstract
OBJECTIVE Pregnant women with COVID-19 are at elevated risk for severe outcomes, but clinical data on management of these patients are limited. Monoclonal antibodies, such as casirivimab plus imdevimab (CAS+IMD), have proven effective in treating non-pregnant adults with COVID-19, prompting further evaluation in pregnant women. METHODS A phase 3 portion of an adaptive, multicentre, randomised, double-blind, placebo-controlled trial evaluated the safety, clinical outcomes, pharmacokinetics and immunogenicity of CAS+IMD (1200 mg or 2400 mg) in the treatment of pregnant outpatients with COVID-19 (NCT04425629). Participants were enrolled between December 2020 and November 2021, prior to the emergence of Omicron-lineage variants against which CAS+IMD is not active. Safety was evaluated in randomised participants who received study drug (n=80); clinical outcomes were evaluated in all randomised participants (n=82). Only two pregnant participants received placebo, limiting conclusions regarding treatment effect. Infants born to pregnant participants were followed for developmental outcomes ≤1 year of age. RESULTS In pregnant participants, CAS+IMD was well tolerated, with no grade ≥2 hypersensitivity or infusion-related reactions reported. There were no participant deaths, and only one COVID-19-related medically attended visit. Although two pregnancies (3%) reported issues in the fetus/neonate, they were confounded by maternal history or considered to be due to an alternate aetiology. No adverse developmental outcomes in infants ≤1 year of age were considered related to in utero exposure to the study drug. CAS+IMD 1200 mg and 2400 mg rapidly and similarly reduced viral loads, with a dose-proportional increase in concentrations of CAS+IMD in serum. Pharmacokinetics were consistent with that reported in the general population. Immunogenicity incidence was low. CONCLUSION CAS+IMD treatment of pregnant outpatients with COVID-19 showed similar safety, clinical outcomes and pharmacokinetic profiles to that observed in non-pregnant adults. There was no evidence of an impact on developmental outcomes in infants ≤1 year of age. TRIAL REGISTRATION NUMBER NCT04425629.
Collapse
Affiliation(s)
| | - Mazhar Thakur
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Samit Ganguly
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Shazia Ali
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Jesse Chao
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | - Jing Xiao
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Yogesh Patel
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | - John D Davis
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Susan C Irvin
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Cynthia Pan
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | | | | | | | | | - Bari Kowal
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | - Rafia Bhore
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | - Edward Cox
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | - Paula Dakin
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Gary A Herman
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| |
Collapse
|
18
|
Kole C, Stefanou Ε, Karvelas N, Schizas D, Toutouzas KP. Acute and Post-Acute COVID-19 Cardiovascular Complications: A Comprehensive Review. Cardiovasc Drugs Ther 2024; 38:1017-1032. [PMID: 37209261 PMCID: PMC10199303 DOI: 10.1007/s10557-023-07465-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 05/22/2023]
Abstract
PURPOSE OF REVIEW The risk of cardiovascular complications due to SARS-CoV-2 are significantly increased within the first 6 months of the infection. Patients with COVID-19 have an increased risk of death, and there is evidence that many may experience a wide range of post-acute cardiovascular complications. Our work aims to provide an update on current clinical aspects of diagnosis and treatment of cardiovascular manifestations during acute and long-term COVID-19. RECENT FINDINGS SARS-CoV-2 has been shown to be associated with increased incidence of cardiovascular complications such as myocardial injury, heart failure, and dysrhythmias, as well as coagulation abnormalities not only during the acute phase but also beyond the first 30 days of the infection, associated with high mortality and poor outcomes. Cardiovascular complications during long-COVID-19 were found regardless of comorbidities such as age, hypertension, and diabetes; nevertheless, these populations remain at high risk for the worst outcomes during post-acute COVID-19. Emphasis should be given to the management of these patients. Treatment with low-dose oral propranolol, a beta blocker, for heart rate management may be considered, since it was found to significantly attenuate tachycardia and improve symptoms in postural tachycardia syndrome, while for patients on ACE inhibitors or angiotensin-receptor blockers (ARBs), under no circumstances should these medications be withdrawn. In addition, in patients at high risk after hospitalization due to COVID-19, thromboprophylaxis with rivaroxaban 10 mg/day for 35 days improved clinical outcomes compared with no extended thromboprophylaxis. In this work we provide a comprehensive review on acute and post-acute COVID-19 cardiovascular complications, symptomatology, and pathophysiology mechanisms. We also discuss therapeutic strategies for these patients during acute and long-term care and highlight populations at risk. Our findings suggest that older patients with risk factors such as hypertension, diabetes, and medical history of vascular disease have worse outcomes during acute SARS-CoV-2 infection and are more likely to develop cardiovascular complications during long-COVID-19.
Collapse
Affiliation(s)
- Christo Kole
- Cardiology Department, Sismanoglio General Hospital of Attica, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Εleni Stefanou
- Artificial Kidney Unit, General Hospital of Messinia, Kalamata, Greece
| | - Nikolaos Karvelas
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | | |
Collapse
|
19
|
Antoniou T, McCormack D, Tadrous M, Gomes T. Alpha-1 adrenergic antagonists and the risk of hospitalization or death in non-hospitalized patients with COVID-19: A population-based study. Fundam Clin Pharmacol 2024; 38:998-1007. [PMID: 38575851 DOI: 10.1111/fcp.13004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/29/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Alpha-1 receptor antagonists may interfere with IL-6 signaling and could therefore be a potential treatment for COVID-19. However, the effectiveness of these drugs in mitigating the risk of clinical deterioration among non-hospitalized patients with COVID-19 is unknown. OBJECTIVES The aim of this study is to examine the association between alpha-1 antagonist exposure and the 30-day risk of a hospital encounter or death in nonhospitalized patients with COVID-19. METHODS We conducted a population-based cohort study of Ontario residents aged 35 years and older who were eligible for public drug coverage and who had a positive test for SARS-CoV-2 between January 1, 2020, and March 1, 2021. We matched each individual receiving an alpha-1 antagonist at the time of their positive test with two non-exposed individuals using propensity scores. Our outcome was a composite of a hospital admission, emergency department visit, or death, 1 to 30 days following the positive test. RESULTS We matched 3289 alpha-1 antagonist exposed patients to 6189 unexposed patients. Overall, there was no difference in the 30-day risk of the primary outcome among patients exposed to alpha-1 antagonists at the time of their diagnosis relative to unexposed individuals (28.8% vs. 28.0%; OR 1.00, 95% CI 0.91 to 1.11). In a secondary analysis, individuals exposed to alpha-1 antagonists had a lower risk of death in the 30 days following a COVID diagnosis (OR 0.79; 95% CI 0.66 to 0.93). CONCLUSION Alpha-1 antagonists did not mitigate the 30-day risk of clinical deterioration in non-hospitalized patients with COVID-19. Our findings do not support the general repurposing of alpha-1 antagonists as a treatment for such patients, although there may be subgroups of patients in whom further research is warranted.
Collapse
Affiliation(s)
- Tony Antoniou
- Department of Family and Community Medicine, Unity Health Toronto, Toronto, Ontario, Canada
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Daniel McCormack
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Mina Tadrous
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Women's College Research Institute, Toronto, Ontario, Canada
| | - Tara Gomes
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Institute for Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Huang Z, Iqbal Z, Zhao Z, Chen X, Mahmmod A, Liu J, Li W, Deng Z. TMEM16 proteins: Ca 2+‑activated chloride channels and phospholipid scramblases as potential drug targets (Review). Int J Mol Med 2024; 54:81. [PMID: 39092585 PMCID: PMC11315658 DOI: 10.3892/ijmm.2024.5405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/06/2024] [Indexed: 08/04/2024] Open
Abstract
TMEM16 proteins, which function as Ca2+‑activated Cl‑ channels are involved in regulating a wide variety of cellular pathways and functions. The modulators of Cl‑ channels can be used for the molecule‑based treatment of respiratory diseases, cystic fibrosis, tumors, cancer, osteoporosis and coronavirus disease 2019. The TMEM16 proteins link Ca2+ signaling, cellular electrical activity and lipid transport. Thus, deciphering these complex regulatory mechanisms may enable a more comprehensive understanding of the physiological functions of the TMEM16 proteins and assist in ascertaining the applicability of these proteins as potential pharmacological targets for the treatment of a range of diseases. The present review examined the structures, functions and characteristics of the different types of TMEM16 proteins, their association with the pathogenesis of various diseases and the applicability of TMEM16 modulator‑based treatment methods.
Collapse
Affiliation(s)
- Zeqi Huang
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zoya Iqbal
- Department of Orthopaedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zhe Zhao
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Xiaoqiang Chen
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Ayesha Mahmmod
- Faculty of Pharmacy, The University of Lahore, Lahore, Punjab 58240, Pakistan
| | - Jianquan Liu
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Wencui Li
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zhiqin Deng
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
21
|
Ximenes Braz B, Cavalcante Meneses G, Bezerra da Silva Junior G, Costa Martins AM, de Souza Mourão Feitosa AF, Cavalcante Lima Chagas G, De Francesco Daher E. Risk factors for mortality in coronavirus disease 2019 patients with silent hypoxemia. Rev Clin Esp 2024; 224:485-493. [PMID: 38945525 DOI: 10.1016/j.rceng.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/02/2024] [Indexed: 07/02/2024]
Abstract
OBJECTIVE To describe the predictors of mortality in hospitalized patients with severe acute respiratory syndrome (SARS) due to COVID-19 presenting with silent hypoxemia. MATERIAL AND METHODS Retrospective cohort study of hospitalized patients with SARS due to COVID-19 and silent hypoxemia at admission, in Brazil, from January to June 2021. The primary outcome of interest was in-hospital death. Multivariable logistic regression analysis was performed. RESULTS Of 46,102 patients, the mean age was 59 ± 16 years, and 41.6% were female. During hospitalization, 13,149 patients died. Compared to survivors, non-survivors were older (mean age, 66 vs. 56 years; P < 0.001), less frequently female (43.6% vs. 40.9%; P < 0.001), and more likely to have comorbidities (74.3% vs. 56.8%; P < 0.001). Non-survivors had higher needs for invasive mechanical ventilation (42.4% vs. 6.6%; P < 0.001) and intensive care unit admission (56.9% vs. 20%; P < 0.001) compared to survivors. In the multivariable regression analysis, advanced age (OR 1.04; 95%CI 1.037-1.04), presence of comorbidities (OR 1.54; 95%CI 1.47-1.62), cough (OR 0.74; 95%CI 0.71-0.79), respiratory distress (OR 1.32; 95%CI 1.26-1.38), and need for non-invasive respiratory support (OR 0.37; 95%CI 0.35-0.40) remained independently associated with death. CONCLUSIONS Advanced age, presence of comorbidities, and respiratory distress were independent risk factors for mortality, while cough and requirement for non-invasive respiratory support were independent protective factors against mortality in hospitalized patients due to SARS due to COVID-19 with silent hypoxemia at presentation.
Collapse
Affiliation(s)
- Beatriz Ximenes Braz
- Post-Graduation Program in Medical Sciences, Department of Internal Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Internal Medicine Department, University of Miami, Miami, FL, United States
| | - Gdayllon Cavalcante Meneses
- Post-Graduation Program in Medical Sciences, Department of Internal Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Clinical and Toxicological Analysis Department, School of Pharmacy, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Geraldo Bezerra da Silva Junior
- Clinical and Toxicological Analysis Department, School of Pharmacy, Federal University of Ceará, Fortaleza, CE, Brazil; Post-Graduation Program in Collective Health, Health Sciences Center, University of Fortaleza - UNIFOR, Fortaleza, CE, Brazil
| | - Alice Maria Costa Martins
- Clinical and Toxicological Analysis Department, School of Pharmacy, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Gabriel Cavalcante Lima Chagas
- Post-Graduation Program in Medical Sciences, Department of Internal Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil.
| | - Elizabeth De Francesco Daher
- Post-Graduation Program in Medical Sciences, Department of Internal Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
22
|
Liu S, Yu C, Tu Q, Zhang Q, Fu Z, Huang Y, He C, Yao L. Bacterial co-infection in COVID-19: a call to stay vigilant. PeerJ 2024; 12:e18041. [PMID: 39308818 PMCID: PMC11416760 DOI: 10.7717/peerj.18041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 08/13/2024] [Indexed: 09/25/2024] Open
Abstract
Co-infection with diverse bacteria is commonly seen in patients infected with the novel coronavirus, SARS-CoV-2. This type of co-infection significantly impacts the occurrence and development of novel coronavirus infection. Bacterial co-pathogens are typically identified in the respiratory system and blood culture, which complicates the diagnosis, treatment, and prognosis of COVID-19, and even exacerbates the severity of disease symptoms and increases mortality rates. However, the status and impact of bacterial co-infections during the COVID-19 pandemic have not been properly studied. Recently, the amount of literature on the co-infection of SARS-CoV-2 and bacteria has gradually increased, enabling a comprehensive discussion on this type of co-infection. In this study, we focus on bacterial infections in the respiratory system and blood of patients with COVID-19 because these infection types significantly affect the severity and mortality of COVID-19. Furthermore, the progression of COVID-19 has markedly elevated the antimicrobial resistance among specific bacteria, such as Klebsiella pneumoniae, in clinical settings including intensive care units (ICUs). Grasping these resistance patterns is pivotal for the optimal utilization and stewardship of antibiotics, including fluoroquinolones. Our study offers insights into these aspects and serves as a fundamental basis for devising effective therapeutic strategies. We primarily sourced our articles from PubMed, ScienceDirect, Scopus, and Google Scholar. We queried these databases using specific search terms related to COVID-19 and its co-infections with bacteria or fungi, and selectively chose relevant articles for inclusion in our review.
Collapse
Affiliation(s)
- Shengbi Liu
- Department of Clinical Laboratory, Guiqian International General Hospital, Guiyang, People’s Republic of China
| | - Chao Yu
- Department of Clinical Laboratory, Guiqian International General Hospital, Guiyang, People’s Republic of China
| | - Qin Tu
- Department of Clinical Laboratory, Guiqian International General Hospital, Guiyang, People’s Republic of China
| | - Qianming Zhang
- Department of Clinical Laboratory, Guiqian International General Hospital, Guiyang, People’s Republic of China
| | - Zuowei Fu
- Department of Clinical Laboratory, Guiqian International General Hospital, Guiyang, People’s Republic of China
| | - Yifeng Huang
- Department of Clinical Laboratory, Guiqian International General Hospital, Guiyang, People’s Republic of China
| | - Chuan He
- Department of Clinical Laboratory, Guiqian International General Hospital, Guiyang, People’s Republic of China
| | - Lei Yao
- Department of Clinical Laboratory, Guiqian International General Hospital, Guiyang, People’s Republic of China
| |
Collapse
|
23
|
Tisch C, Xourgia E, Exadaktylos A, Ziaka M. Potential use of sodium glucose co-transporter 2 inhibitors during acute illness: a systematic review based on COVID-19. Endocrine 2024; 85:660-675. [PMID: 38448675 PMCID: PMC11291544 DOI: 10.1007/s12020-024-03758-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
OBJECTIVE SGLT-2i are increasingly recognized for their benefits in patients with cardiometabolic risk factors. Additionally, emerging evidence suggests potential applications in acute illnesses, including COVID-19. This systematic review aims to evaluate the effects of SGLT-2i in patients facing acute illness, particularly focusing on SARS-CoV-2 infection. METHODS Following PRISMA guidelines, a systematic search of PubMed, Scopus, medRxiv, Research Square, and Google Scholar identified 22 studies meeting inclusion criteria, including randomized controlled trials and observational studies. Data extraction and quality assessment were conducted independently. RESULTS Out of the 22 studies included in the review, six reported reduced mortality in DM-2 patients taking SGLT-2i, while two found a decreased risk of hospitalization. Moreover, one study demonstrated a lower in-hospital mortality rate in DM-2 patients under combined therapy of metformin plus SGLT-2i. However, three studies showed a neutral effect on the risk of hospitalization. No increased risk of developing COVID-19 was associated with SGLT-2i use in DM-2 patients. Prior use of SGLT-2i was not associated with ICU admission and need for MV. The risk of acute kidney injury showed variability, with inconsistent evidence regarding diabetic ketoacidosis. CONCLUSION Our systematic review reveals mixed findings on the efficacy of SGLT-2i use in COVID-19 patients with cardiometabolic risk factors. While some studies suggest potential benefits in reducing mortality and hospitalizations, others report inconclusive results. Further research is needed to clarify optimal usage and mitigate associated risks, emphasizing caution in clinical interpretation.
Collapse
Affiliation(s)
- Carmen Tisch
- Department of Internal Medicine, Thun General Hospital, Thun, Switzerland
| | - Eleni Xourgia
- Department of Cardiology, Inselspital, University Hospital, University of Bern, 3008, Bern, Switzerland
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Mairi Ziaka
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
24
|
Lazar SD, Stoenescu AF, Popescu CP, Florescu S, Vancea G, Calistru P. The Interplay of Chronic Hepatitis C and COVID-19: Implications for Prognosis and Treatment. Cureus 2024; 16:e66639. [PMID: 39258096 PMCID: PMC11386413 DOI: 10.7759/cureus.66639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Chronic hepatitis C (CHC) remains a significant public health concern due to both hepatic and extrahepatic manifestations associated with substantial morbidity and mortality. The emergence of SARS-CoV-2 has raised concerns about the outcomes of COVID-19 in CHC patients. Method We conducted a retrospective analysis of patients with CHC and SARS-CoV-2 infection admitted to a tertiary care hospital between 2020 and 2023. We performed a global analysis of the entire batch of patients and, later, we evaluated the patients according to the severity of the SARS-CoV-2 infection Results The cohort included 89 patients (63 females, 26 males) with a median age of 65 years. Most patients were hospitalized in 2021. Common clinical manifestations included fever, cough, digestive symptoms, and headache. The most frequent comorbidities were renal disease, thyroid disorders, and cancer. Univariate logistic regression analysis identified older age, hospitalization in 2021, and respiratory failure as risk factors for severe COVID-19. Elevated lactate dehydrogenase levels were also associated with an increased risk of severe COVID-19. Regarding CHC, detectable hepatitis C virus viremia was associated with more severe liver disease (p<0.01). Conclusion Patients with CHC and SARS-CoV-2 infection have a substantial risk of severe outcomes. Early identification and management of these patients are crucial to improve their prognosis.
Collapse
Affiliation(s)
- Stefan D Lazar
- Infectious Diseases and Tropical Medicine, Dr Victor Babes Hospital of Infectious and Tropical Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Andreea F Stoenescu
- Infectious Diseases and Tropical Medicine, Dr Victor Babes Hospital of Infectious and Tropical Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Corneliu Petru Popescu
- Infectious Diseases and Tropical Medicine, Dr Victor Babes Hospital of Infectious and Tropical Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Simin Florescu
- Infectious Diseases and Tropical Medicine, Dr Victor Babes Hospital of Infectious and Tropical Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Geta Vancea
- Infectious Diseases and Tropical Medicine, Dr Victor Babes Hospital of Infectious and Tropical Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Petre Calistru
- Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| |
Collapse
|
25
|
Pradhevi L, Soegiarto G, Wulandari L, Lusida MA, Saefudin RP, Vincent A. More severe comorbidities, advanced age, and incomplete vaccination increase the risk of COVID-19 mortality. NARRA J 2024; 4:e949. [PMID: 39280314 PMCID: PMC11391969 DOI: 10.52225/narra.v4i2.949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024]
Abstract
Numerous studies have stated that comorbidities are risk factors for coronavirus disease 2019 (COVID-19) mortality, but few have considered the severity or stage of these comorbidities. The aim of this study was to determine the association between the severity of comorbidity, age, and number of COVID-19 vaccinations with COVID-19 mortality. This case-control study was conducted from July 2021 until December 2022 at the Dr. Soetomo General Academic Hospital, Surabaya, Indonesia. The patients were divided into non-survived patients (case group) and survived patients (control group). The inclusion criteria for cases were adult patients hospitalized with confirmed COVID-19, based on reverse transcriptase-polymerase chain reaction (RT-PCR) testing of nasopharyngeal swabs. Using total sampling, 1,046 confirmed COVID-19 patients, which consisted of 450 (43%) non-survived patients and 596 (57%) survived patients, were included. The most common comorbidity was diabetes mellitus (DM) (82.7%), chronic kidney disease (CKD) (43%), hypertension (25.7%), and obesity (23.6%). Our multivariate analysis indicated that older age (aOR: 1.03; 95%CI: 1.02-1.04, p<0.001), male sex (aOR: 1.29; 95%CI: 1.11- 2.00, p=0.007), severe COVID-19 at first admission (aOR: 3.13; 95%CI: 2.08-4.73, p<0.001), having pneumonia (aOR: 1.99; 95%CI: 1.21-3.33, p=0.005), poorly controlled DM with HbA1c≥9% (aOR: 2.90; 95%CI: 1.72-4.89, p<0.001), severe obesity with body mass index (BMI)≥30 (OR: 2.90; 95%CI: 1.72-4.89, p<0.001), hypertension stage 2 (aOR: 1.99; 95%CI: 1.12-3.53, p=0.019) or stage 3 (aOR: 6.59; 95%CI: 2.39-18.17, p<0.001), CKD stage 3 (aOR: 2.50; 95%CI: 1.36-4.59, p=0.003), stage 4 (aOR: 5.47; 95%CI: 2.18-13.69, p<0.001) or stage 5 (aOR: 1.71; 95%CI: 1.04-2.81, p=0.036), and having chronic lung disease (aOR: 3.08; 95%CI: 1.22-7.77, p=0.017) significantly increased the risk of COVID-19 mortality. In contrast, COVID-19 vaccination reduced the risk of COVID-19-associated death. This study highlights that more severe comorbidities, advanced age, and incomplete vaccination were associated with COVID-19 mortality.
Collapse
Affiliation(s)
- Lukita Pradhevi
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Gatot Soegiarto
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Laksmi Wulandari
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Pulmonology and Respiratory Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Michael Ap Lusida
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Rendra P Saefudin
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Agustinus Vincent
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
26
|
Matamalas JT, Chelvanambi S, Decano JL, França RF, Halu A, Santinelli-Pestana DV, Aikawa E, Malhotra R, Aikawa M. Obesity and age are transmission risk factors for SARS-CoV-2 infection among exposed individuals. PNAS NEXUS 2024; 3:pgae294. [PMID: 39192848 PMCID: PMC11348562 DOI: 10.1093/pnasnexus/pgae294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/07/2024] [Indexed: 08/29/2024]
Abstract
The coronavirus disease (COVID-19) pandemic has occurred in Massachusetts in multiple waves led by a series of emerging variants. While the evidence has linked obesity with severe symptoms of COVID-19, the effect of obesity on susceptibility to SARS-CoV-2 infection remains unclear. Identification of intrinsic factors, which increase the likelihood of exposed individuals succumbing to productive SARS-CoV-2 infection could help plan mitigation efforts to curb the illness. We aim to investigate whether obese individuals have a higher susceptibility to developing productive SARS-CoV-2 infection given comparable exposure to nonobese individuals. This case-control study leveraged data from the Mass General Brigham's (MGB) electronic medical records (EMR), containing 687,813 patients, to determine whether obesity at any age increases the proportion of infections. We used PCR results of 72,613 subjects who tested positive to SARS-CoV-2 or declared exposure to the virus independently of the result of the test. For this study, we defined susceptibility as the likelihood of testing positive upon suspected exposure. We demonstrate evidence that SARS-CoV-2 exposed obese individuals were more prone to become COVID positive than nonobese individuals [adjusted odds ratio = 1.34 (95% CI: 1.29-1.39)]. Temporal analysis showed significantly increased susceptibility in obese individuals across the duration of the pandemic in Massachusetts. Obese exposed individuals are at a higher risk of getting infected with SARS-CoV-2. This indicates that obesity is not only a risk factor for worsened outcomes but also increases the risk for infection upon exposure. Identifying such populations early will be crucial for curbing the spread of this infectious disease.
Collapse
Affiliation(s)
- Joan T Matamalas
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Sarvesh Chelvanambi
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Julius L Decano
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Raony F França
- Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455 - Cerqueira César, São Paulo, SP 01246-903, Brazil
| | - Arda Halu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Diego V Santinelli-Pestana
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Elena Aikawa
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
- Department of Medicine, Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Rajeev Malhotra
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Masanori Aikawa
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
- Department of Medicine, Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
27
|
Venkatraman S, Salinero JMG, Scheinfeld A, Houghton S, Redmond D, Safford M, Rajan M. Clusters of long COVID among patients hospitalized for COVID-19 in New York City. BMC Public Health 2024; 24:1994. [PMID: 39061026 PMCID: PMC11282800 DOI: 10.1186/s12889-024-19379-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Recent studies have demonstrated that individuals hospitalized due to COVID-19 can be affected by "long-COVID" symptoms for as long as one year after discharge. OBJECTIVES Our study objective is to identify data-driven clusters of patients using a novel, unsupervised machine learning technique. METHODS The study uses data from 437 patients hospitalized in New York City between March 3rd and May 15th of 2020. The data used was abstracted from medical records and collected from a follow-up survey for up to one-year post-hospitalization. Hospitalization data included demographics, comorbidities, and in-hospital complications. The survey collected long-COVID symptoms, and information on general health, social isolation, and loneliness. To perform the analysis, we created a graph by projecting the data onto eight principal components (PCs) and running the K-nearest neighbors algorithm. We then used Louvain's algorithm to partition this graph into non-overlapping clusters. RESULTS The cluster analysis produced four clusters with distinct health and social connectivity patterns. The first cluster (n = 141) consisted of patients with both long-COVID neurological symptoms (74%) and social isolation/loneliness. The second cluster (n = 137) consisted of healthy patients who were also more socially connected and not lonely. The third cluster (n = 96) contained patients with neurological symptoms who were socially connected but lonely, and the fourth cluster (n = 63) consisted entirely of patients who had traumatic COVID hospitalization, were intubated, suffered symptoms, but were socially connected and experienced recovery. CONCLUSION The cluster analysis identified social isolation and loneliness as important features associated with long-COVID symptoms and recovery after hospitalization. It also confirms that social isolation and loneliness, though connected, are not necessarily the same. Physicians need to be aware of how social characteristics relate to long-COVID and patient's ability to cope with the resulting symptoms.
Collapse
Affiliation(s)
- Sara Venkatraman
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medicine, 420 E 70th street LH 348, New York, NY, USA
- Department of Statistics and Data Science, Cornell University, Ithaca, NY, USA
| | - Jesus Maria Gomez Salinero
- Division of Regenerative Medicine, Department of Medicine, Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell Medicine, New York, NY, USA
| | - Adina Scheinfeld
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Sean Houghton
- Division of Regenerative Medicine, Department of Medicine, Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Department of Medicine, Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell Medicine, New York, NY, USA
| | - Monika Safford
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medicine, 420 E 70th street LH 348, New York, NY, USA
| | - Mangala Rajan
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medicine, 420 E 70th street LH 348, New York, NY, USA.
| |
Collapse
|
28
|
Douglas ME, Mamawala M, Brown S, Powers MB, Warren AM. Associations of mood symptoms and body mass among individuals with COVID-19. Proc AMIA Symp 2024; 37:755-762. [PMID: 39165826 PMCID: PMC11332635 DOI: 10.1080/08998280.2024.2358294] [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: 11/30/2023] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 08/22/2024] Open
Abstract
Introduction The COVID-19 pandemic has had a significant impact on mental health, and individuals with certain preexisting morbidities-such as obesity-may have disproportionately experienced pandemic-related stress. We aimed to examine whether being obese or overweight was associated with worse mood and trauma among those infected with COVID-19. Methods Data were collected as part of a larger longitudinal survey study (n = 591) examining the psychological impact of COVID-19. Complete baseline data for those who reported testing positive from April 2020 through June 2021 were examined. Results and conclusions Body mass index was correlated with depression (r = 0.13, P < .01) and generalized anxiety disorder (r = 0.10, P = .02), but not with posttraumatic stress disorder (r = 0.06, P = .16). Multivariable analyses revealed that body mass index was no longer a unique predictor, except for those with a body mass index >40 kg/m2, suggesting that other sociodemographic factors, not obesity, were driving mood symptoms for those infected with COVID-19.
Collapse
Affiliation(s)
| | | | - Shakeira Brown
- Baylor Scott & White Research Institute, Dallas, Texas, USA
| | - Mark B. Powers
- Baylor Scott & White Research Institute, Dallas, Texas, USA
| | | |
Collapse
|
29
|
Telang MM, Waghaye S, Muthusamy E, Choudhary S, Alrais ZF, Kasim F, Khatib KI. Association of obesity and mortality in patients with COVID-19 acute respiratory distress syndrome: A retrospective cohort study. Int J Crit Illn Inj Sci 2024; 14:153-159. [PMID: 39512557 PMCID: PMC11540193 DOI: 10.4103/ijciis.ijciis_27_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 11/15/2024] Open
Abstract
Background Chronic conditions such as obesity are associated with adverse outcomes in coronavirus disease 2019 (COVID-19) acute respiratory distress syndrome (ARDS) patients. The aim of our study was to evaluate the relationship between BMI and outcomes in critically ill patients with COVID-19 ARDS. Methods A retrospective study including all patients with COVID-19 and ARDS on mechanical ventilation admitted to the intensive care unit (ICU) over 2 years. Patients with obesity (BMI ≥30 kg/m2) were compared with those without obesity (BMI >18.5 up to 29.9 kg/m2). Outcomes compared were primary (mortality, duration of mechanical ventilation, and length of ICU stay) and secondary complications during the ICU course (inotrope requirement, acute kidney injury [AKI] requiring renal replacement therapy [RRT], and bloodstream and urinary tract infections). Results One hundred and eight patients were included in the study. The mean age of patients was 52 years, and 94 (87%) patients were males. As compared to COVID-19 ARDS patients without obesity, COVID-19 patients with obesity were more prone to develop complications like AKI, necessitating continuous RRT (P = 0.005). There was no significant difference in other complications between the two groups (all P > 0.05). There was no increased mortality in these obese patients (P = 0.056). In these patients with obesity, those who also had ischemic heart disease had an increased likelihood of mortality (P = 0.036). Conclusion Our study concludes that patients with COVID-19 ARDS who are obese are not at higher risk of mortality and more likely to develop renal complications. When these patients develop cardiac complications or bloodstream infections, they have a significantly higher risk of mortality.
Collapse
Affiliation(s)
- Madhavi Mahesh Telang
- Rashid Hospital and Emergency Trauma Centre, Intensive Care Unit, Dubai, United Arab Emirates
| | - Samrat Waghaye
- Rashid Hospital and Emergency Trauma Centre, Intensive Care Unit, Dubai, United Arab Emirates
| | - Elangho Muthusamy
- Rashid Hospital and Emergency Trauma Centre, Intensive Care Unit, Dubai, United Arab Emirates
| | - Sunil Choudhary
- Rashid Hospital and Emergency Trauma Centre, Intensive Care Unit, Dubai, United Arab Emirates
| | - Zeyad Faoor Alrais
- Rashid Hospital and Emergency Trauma Centre, Intensive Care Unit, Dubai, United Arab Emirates
| | - Fathima Kasim
- Rashid Hospital and Emergency Trauma Centre, Intensive Care Unit, Dubai, United Arab Emirates
| | | |
Collapse
|
30
|
Zhang R, Wei R, Yuan Y, Li N, Hu Y, Chan KH, Hung IFN, Tse HF. Human-induced pluripotent stem cell-derived hepatocyte platform in modeling of SARS-CoV-2 infection. JGH Open 2024; 8:e13039. [PMID: 39006099 PMCID: PMC11239974 DOI: 10.1002/jgh3.13039] [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: 06/27/2023] [Revised: 12/31/2023] [Accepted: 02/08/2024] [Indexed: 07/16/2024]
Abstract
Background and Aim Currently, SARS-CoV-2 is still spreading rapidly and globally. A large proportion of patients with COVID-19 developed liver injuries. The human-induced pluripotent stem cell (iPSC)-derived hepatocytes recapitulate primary human hepatocytes and have been widely used in studies of liver diseases. Methods To explore the susceptibility of hepatocytes to SARS-CoV-2, we differentiated iPSCs to functional hepatocytes and tried infecting them with different MOI (1, 0.1, 0.01) of SARS-CoV-2. Results The iPSC-derived hepatocytes are highly susceptible to virus infection, even at 0.01 MOI. Other than the ancestral strain, iHeps also support the replication of SARS-CoV-2 variants including alpha, beta, theta, and delta. More interestingly, the ACE2 expression significantly upregulated after infection, suggesting a vicious cycle between virus infection and liver injury. Conclusions The iPSC-derived hepatocytes can support the replication of SARS-CoV-2, and this platform could be used to investigate the SARS-CoV-2 hepatotropism and hepatic pathogenic mechanisms.
Collapse
Affiliation(s)
- Ruiqi Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Rui Wei
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
- Department of Gastroenterology and Hepatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences) Southern Medical University Guangzhou China
- Center for Translational Stem Cell Biology Hong Kong SAR China
| | - Yangyang Yuan
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
- Center for Translational Stem Cell Biology Hong Kong SAR China
| | - Na Li
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Yang Hu
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Kwok-Hung Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Hung-Fat Tse
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
- Center for Translational Stem Cell Biology Hong Kong SAR China
- Cardiac and Vascular Center Hong Kong University Shenzhen Hospital Shenzhen China
- Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine The University of Hong Kong Hong Kong SAR China
| |
Collapse
|
31
|
Lei Z, Liu D, Li M, Xian D, Fan S. Vaccination intentions of hypertensive Chinese individuals during the COVID-19 epidemic: a structural equation modeling study. BMC Infect Dis 2024; 24:642. [PMID: 38926657 PMCID: PMC11201319 DOI: 10.1186/s12879-024-09480-0] [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: 06/09/2023] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVE Given the high prevalence of hypertension among Chinese adults, this population is at a significantly increased risk of severe COVID-19 complications. The purpose of this study is to assess the willingness of Chinese hypertensive adults to receive the COVID-19 vaccine and to identify the diverse factors that shape their vaccination decisions. METHODS Sampling was conducted utilizing multistage stratified random sampling, and ultimately, a total of 886 adult hypertensive patients from Luzhou City in Southwest China were included in this study. The questionnaire design was based on the Theory of Planned Behaviour and was used to investigate their willingness to be vaccinated with COVID-19. Structural equation modeling was employed for data analysis. RESULTS The results showed that 75.6% of hypertensive individuals were willing to receive COVID-19 vaccination. The structural equation modeling revealed that Subjective Norms (path coefficient = 0.361, CR = 8.049, P < 0.001) and Attitudes (path coefficient = 0.253, CR = 4.447, P < 0.001) had positive effects on vaccination willingness, while Perceived Behavioral Control (path coefficient=-0.004, CR=-0.127, P = 0.899) had no significant impact on Behavioral Attitudes. Mediation analysis indicated that Knowledge (indirect path coefficient = 0.032, LLCI = 0.014, ULCI = 0.058), Risk Perception (indirect path coefficient = 0.077, LLCI = 0.038, ULCI = 0.124), and Subjective Norms (indirect path coefficient = 0.044, LLCI = 0.019, ULCI = 0.087) significantly influenced vaccination willingness through Attitudes as a mediating factor. CONCLUSION The willingness of hypertensive individuals to receive the COVID-19 vaccination is not satisfactory. The Theory of Planned Behavior provides valuable insights into understanding their vaccination intentions. Efforts should be concentrated on enhancing the subjective norms, attitudes, and knowledge about vaccination of hypertensive patients.
Collapse
Affiliation(s)
- Zhi Lei
- Department of Chronic Disease Control and Prevention, Luzhou Center for Disease Control and Prevention, Luzhou, China
- Administrative Office, Luzhou People's Hospital, Luzhou, China
| | - Dongyang Liu
- School of Public Health, Southwest Medical University, Luzhou, China
| | - Minghui Li
- Department of Human Resource, Sichuan Provincial Center for Disease Control and Prevention, Chengdu, China
| | - Deqiang Xian
- Administrative Office, Luzhou People's Hospital, Luzhou, China.
| | - Song Fan
- School of Public Health, Southwest Medical University, Luzhou, China.
| |
Collapse
|
32
|
Lima TE, Ferraz MVF, Brito CAA, Ximenes PB, Mariz CA, Braga C, Wallau GL, Viana IFT, Lins RD. Determination of prognostic markers for COVID-19 disease severity using routine blood tests and machine learning. AN ACAD BRAS CIENC 2024; 96:e20230894. [PMID: 38922277 DOI: 10.1590/0001-376520242023089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 02/22/2024] [Indexed: 06/27/2024] Open
Abstract
The need for the identification of risk factors associated to COVID-19 disease severity remains urgent. Patients' care and resource allocation can be potentially different and are defined based on the current classification of disease severity. This classification is based on the analysis of clinical parameters and routine blood tests, which are not standardized across the globe. Some laboratory test alterations have been associated to COVID-19 severity, although these data are conflicting partly due to the different methodologies used across different studies. This study aimed to construct and validate a disease severity prediction model using machine learning (ML). Seventy-two patients admitted to a Brazilian hospital and diagnosed with COVID-19 through RT-PCR and/or ELISA, and with varying degrees of disease severity, were included in the study. Their electronic medical records and the results from daily blood tests were used to develop a ML model to predict disease severity. Using the above data set, a combination of five laboratorial biomarkers was identified as accurate predictors of COVID-19 severe disease with a ROC-AUC of 0.80 ± 0.13. Those biomarkers included prothrombin activity, ferritin, serum iron, ATTP and monocytes. The application of the devised ML model may help rationalize clinical decision and care.
Collapse
Affiliation(s)
- Tayná E Lima
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Departamento de Virologia, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Matheus V F Ferraz
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Departamento de Virologia, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
- Universidade Federal de Pernambuco, Departamento de Química Fundamental, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-560 Recife, PE, Brazil
| | - Carlos A A Brito
- Universidade Federal de Pernambuco, Hospital das Clínicas, Av. Professor Moraes Rego, 1235, Cidade Universitária, 50670-901 Recife, PE, Brazil
| | - Pamella B Ximenes
- Hospital dos Servidores Públicos do Estado de Pernambuco, Av. Conselheiro Rosa e Silva, s/n, Espinheiro, 52020-020 Recife, PE, Brazil
| | - Carolline A Mariz
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Departamento de Parasitologia, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Cynthia Braga
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Departamento de Parasitologia, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Gabriel L Wallau
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Departamento de Entomologia, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Isabelle F T Viana
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Departamento de Virologia, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Roberto D Lins
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Departamento de Virologia, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| |
Collapse
|
33
|
Hussain SS, Libby EF, Lever JEP, Tipper JL, Phillips SE, Mazur M, Li Q, Campos-Gómez J, Harrod KS, Rowe SM. ACE-2 Blockade & TMPRSS2 Inhibition Mitigate SARS-CoV-2 Severity Following Cigarette Smoke Exposure in Airway Epithelial Cells In Vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.23.600238. [PMID: 38979208 PMCID: PMC11230175 DOI: 10.1101/2024.06.23.600238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Cigarette smoking is associated with COVID-19 prevalence and severity, but the mechanistic basis for how smoking alters SARS-CoV-2 pathogenesis is unknown. A potential explanation is that smoking alters the expression of the SARS-CoV-2 cellular receptor and point of entry, angiotensin converting enzyme-2 (ACE-2), and its cofactors including transmembrane protease serine 2 (TMPRSS2). We investigated the impact of cigarette smoking on the expression of ACE-2, TMPRSS2, and other known cofactors of SARS-CoV-2 infection and the resultant effects on infection severity in vitro. Cigarette smoke extract (CSE) exposure increased ACE-2 and TMPRSS2 mRNA expression compared to air control in ferret airway cells, Calu-3 cells, and primary human bronchial epithelial (HBE) cells derived from normal and COPD donors. CSE-exposed ferret airway cells inoculated with SARS-CoV-2 had a significantly higher intracellular viral load versus vehicle-exposed cells. Likewise, CSE-exposure increased both SARS-CoV-2 intracellular viral load and viral replication in both normal and COPD HBE cells over vehicle control. Apoptosis was increased in CSE-exposed, SARS-CoV-2-infected HBE cells. Knockdown of ACE-2 via an antisense oligonucleotide (ASO) reduced SARS-CoV-2 viral load and infection in CSE-exposed ferret airway cells that was augmented by co-administration of camostat mesylate to block TMPRSS2 activity. Smoking increases SARS-CoV-2 infection via upregulation of ACE2 and TMPRSS2.
Collapse
|
34
|
Raheem MA, Rahim MA, Gul I, Reyad-Ul-Ferdous M, Zhang CY, Yu D, Pandey V, Du K, Wang R, Han S, Han Y, Qin P. COVID-19: Post infection implications in different age groups, mechanism, diagnosis, effective prevention, treatment, and recommendations. Life Sci 2024:122861. [PMID: 38925222 DOI: 10.1016/j.lfs.2024.122861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
SARS-CoV-2 is a highly contagious pathogen that predominantly caused the COVID-19 pandemic. The persistent effects of COVID-19 are defined as an inflammatory or host response to the virus that begins four weeks after initial infection and persists for an undetermined length of time. Chronic effects are more harmful than acute ones thus, this review explored the long-term effects of the virus on various human organs, including the pulmonary, cardiovascular, and neurological, reproductive, gastrointestinal, musculoskeletal, endocrine, and lymphoid systems and found that SARS-CoV-2 adversely affects these organs of older adults. Regarding diagnosis, the RT-PCR is a gold standard method of diagnosing COVID-19; however, it requires specialized equipment and personnel for performing assays and a long time for results production. Therefore, to overcome these limitations, artificial intelligence employed in imaging and microfluidics technologies is the most promising in diagnosing COVID-19. Pharmacological and non-pharmacological strategies are the most effective treatment for reducing the persistent impacts of COVID-19 by providing immunity to post-COVID-19 patients by reducing cytokine release syndrome, improving the T cell response, and increasing the circulation of activated natural killer and CD8 T cells in blood and tissues, which ultimately reduces fever, nausea, fatigue, and muscle weakness and pain. Vaccines such as inactivated viral, live attenuated viral, protein subunit, viral vectored, mRNA, DNA, or nanoparticle vaccines significantly reduce the adverse long-term virus effects in post-COVID-19 patients; however, no vaccine was reported to provide lifetime protection against COVID-19; consequently, protective measures such as physical separation, mask use, and hand cleansing are promising strategies. This review provides a comprehensive knowledge of the persistent effects of COVID-19 on people of varying ages, as well as diagnosis, treatment, vaccination, and future preventative measures against the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Muhammad Akmal Raheem
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Muhammad Ajwad Rahim
- College of Animal Science and Technology, Ahnui Agricultural University, Hefei, PR China
| | - Ijaz Gul
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Md Reyad-Ul-Ferdous
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Can Yang Zhang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Dongmei Yu
- School of Mechanical, Electrical & Information Engineering, Shandong University
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Ke Du
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA
| | - Runming Wang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Sanyang Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Yuxing Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China
| | - Peiwu Qin
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Center of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, Guangdong Province 518055, PR China.
| |
Collapse
|
35
|
Carvajal JJ, García-Castillo V, Cuellar SV, Campillay-Véliz CP, Salazar-Ardiles C, Avellaneda AM, Muñoz CA, Retamal-Díaz A, Bueno SM, González PA, Kalergis AM, Lay MK. New insights into the pathogenesis of SARS-CoV-2 during and after the COVID-19 pandemic. Front Immunol 2024; 15:1363572. [PMID: 38911850 PMCID: PMC11190347 DOI: 10.3389/fimmu.2024.1363572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 04/24/2024] [Indexed: 06/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the respiratory distress condition known as COVID-19. This disease broadly affects several physiological systems, including the gastrointestinal, renal, and central nervous (CNS) systems, significantly influencing the patient's overall quality of life. Additionally, numerous risk factors have been suggested, including gender, body weight, age, metabolic status, renal health, preexisting cardiomyopathies, and inflammatory conditions. Despite advances in understanding the genome and pathophysiological ramifications of COVID-19, its precise origins remain elusive. SARS-CoV-2 interacts with a receptor-binding domain within angiotensin-converting enzyme 2 (ACE2). This receptor is expressed in various organs of different species, including humans, with different abundance. Although COVID-19 has multiorgan manifestations, the main pathologies occur in the lung, including pulmonary fibrosis, respiratory failure, pulmonary embolism, and secondary bacterial pneumonia. In the post-COVID-19 period, different sequelae may occur, which may have various causes, including the direct action of the virus, alteration of the immune response, and metabolic alterations during infection, among others. Recognizing the serious adverse health effects associated with COVID-19, it becomes imperative to comprehensively elucidate and discuss the existing evidence surrounding this viral infection, including those related to the pathophysiological effects of the disease and the subsequent consequences. This review aims to contribute to a comprehensive understanding of the impact of COVID-19 and its long-term effects on human health.
Collapse
Affiliation(s)
- Jonatan J. Carvajal
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Valeria García-Castillo
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Shelsy V. Cuellar
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | | | - Camila Salazar-Ardiles
- Center for Research in Physiology and Altitude Medicine (FIMEDALT), Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Andrea M. Avellaneda
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Department of Basic Sciences, Faculty of Sciences, Universidad Santo Tomás, Antofagasta, Chile
| | - Christian A. Muñoz
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Angello Retamal-Díaz
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| |
Collapse
|
36
|
Bobot M, Heim X, Max H, Boucraut J, Simeone P, Stein C, Velly L, Bruder N, Forel JM, Hraiech S, Guervilly C, Carvelli J, Gainnier M, Mège JL, Chopinet S, Jourde-Chiche N, Papazian L, Burtey S. Prospective Multicenter Study on Early Proximal Tubular Injury in COVID-19-Related Acute Respiratory Distress Syndrome. Kidney Int Rep 2024; 9:1641-1653. [PMID: 38899195 PMCID: PMC11184390 DOI: 10.1016/j.ekir.2024.03.011] [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: 10/31/2023] [Revised: 02/27/2024] [Accepted: 03/11/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction During COVID-19, renal impairment is associated with poor prognosis in intensive care unit (ICU). We aimed to assess the existence and incidence of early renal dysfunction and its prognostic value in patients with COVID-19-related acute respiratory distress syndrome (ARDS). Methods In this prospective multicenter study, patients aged over 18 years with invasive mechanical ventilation (MV) for ARDS were enrolled in 3 ICUs. Precise evaluation of renal dysfunction markers, including urinary protein electrophoresis (UPE) and quantification, was performed within 24 hours after MV onset. Results From March 2020 to December 2021, 135 patients were enrolled as follows: 100 with COVID-19 ARDS and 35 with non-COVID-19 ARDS. UPE found more tubular dysfunction in patients with COVID-19 (68% vs. 21.4%, P < 0.0001) and more normal profiles in patients without COVID-19 (65.0% vs. 11.2%, P = 0.0003). Patients with COVID-19 significantly displayed early urinary leakage of tubular proteins such as beta-2-microglobulin (ß2m) and free light chains, tended to display acute kidney injury (AKI) more frequently (51.0% vs. 34.3%, P = 0.088), had longer MV (20 vs. 9 days, P < 0.0001) and longer ICU stay (26 vs. 15 days, P < 0.0001). In COVID-19 ARDS, leakage of free lambda light chain was associated with the onset of Kidney Disease: Improving Global Outcomes (KDIGO) ≥2 AKI (odds ratio [OR]: 1.014, 95% confidence interval [CI] 1.003-1.025, P = 0.011). Conclusion Patients with COVID-19-related ARDS display a proximal tubular dysfunction before the onset of AKI, which predicts AKI. Proximal tubular damage seems an important mechanism of COVID-19-induced nephropathy. Analysis of urinary proteins is a reliable noninvasive tool to assess proximal tubular dysfunction in the ICU.
Collapse
Affiliation(s)
- Mickaël Bobot
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, AP-HM, Marseille, France
- INSERM, INRAE, C2VN, Aix-Marseille Université, Marseille, France
- CERIMED, Aix-Marseille Université, Marseille, France
| | - Xavier Heim
- INSERM, INRAE, C2VN, Aix-Marseille Université, Marseille, France
- Laboratoire d’Immunologie, Hôpital de la Conception, AP-HM, France
| | - Howard Max
- Département d’Anesthésie-Réanimation, AP-HM, CHU Timone, Aix-Marseille Université, Marseille, France
| | - José Boucraut
- Département d’Anesthésie-Réanimation, AP-HM, CHU Timone, Aix-Marseille Université, Marseille, France
- INT UMR CNRS 7286, Aix-Marseille Université, Marseille, France
| | - Pierre Simeone
- Département d’Anesthésie-Réanimation, AP-HM, CHU Timone, Aix-Marseille Université, Marseille, France
- CNRS, Institut des Neurosciences de la Timone, UMR7289, Marseille, France
| | - Claire Stein
- Assistance Publique - Hôpitaux de Marseille, Hôpital Nord, Médecine Intensive Réanimation; Centre d'Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, France
| | - Lionel Velly
- Département d’Anesthésie-Réanimation, AP-HM, CHU Timone, Aix-Marseille Université, Marseille, France
- CNRS, Institut des Neurosciences de la Timone, UMR7289, Marseille, France
| | - Nicolas Bruder
- Département d’Anesthésie-Réanimation, AP-HM, CHU Timone, Aix-Marseille Université, Marseille, France
| | - Jean-Marie Forel
- Assistance Publique - Hôpitaux de Marseille, Hôpital Nord, Médecine Intensive Réanimation; Centre d'Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, France
| | - Sami Hraiech
- Assistance Publique - Hôpitaux de Marseille, Hôpital Nord, Médecine Intensive Réanimation; Centre d'Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, France
| | - Christophe Guervilly
- Assistance Publique - Hôpitaux de Marseille, Hôpital Nord, Médecine Intensive Réanimation; Centre d'Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, France
| | - Julien Carvelli
- Service de Réanimation et Surveillance Continue, Hôpital de la Timone, AP-HM, Marseille, France
| | - Marc Gainnier
- INSERM, INRAE, C2VN, Aix-Marseille Université, Marseille, France
- Service de Réanimation et Surveillance Continue, Hôpital de la Timone, AP-HM, Marseille, France
| | - Jean-Louis Mège
- Laboratoire d’Immunologie, Hôpital de la Conception, AP-HM, France
| | - Sophie Chopinet
- Service de Chirurgie Générale et Transplantation Hépatique, Hôpital de la Timone, LIIE, CERIMED, Aix-Marseille Université, Marseille, France
| | - Noémie Jourde-Chiche
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, AP-HM, Marseille, France
- INSERM, INRAE, C2VN, Aix-Marseille Université, Marseille, France
| | - Laurent Papazian
- Assistance Publique - Hôpitaux de Marseille, Hôpital Nord, Médecine Intensive Réanimation; Centre d'Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, France
| | - Stéphane Burtey
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, AP-HM, Marseille, France
- INSERM, INRAE, C2VN, Aix-Marseille Université, Marseille, France
| |
Collapse
|
37
|
Qadar SMZ, Naz H, Shamim S, Hashim F, Ahmed S, Kumar Mehraj S. Prevalence of Obesity and its Effects in Patients With COVID-19: A Systematic Review and Meta-analysis. Hosp Pharm 2024; 59:341-348. [PMID: 38764990 PMCID: PMC11097928 DOI: 10.1177/00185787231220318] [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: 05/21/2024]
Abstract
Background: Coronavirus disease 2019 (COVID-19) is an emerging infectious disease worldwide. Obesity has been proven to increase the susceptibility of an individual to infections, but the relationship between obesity and COVID-19 is still unclear. This study aimed to conduct a systematic review and meta-analysis of the prevalence of obesity and its effects in patients with COVID-19. Methods: Web of Science, PubMed and Embase were searched for English language studies up to May 22, 2020. We used a random or fixed-effects model to calculate pooled prevalence rates and odds ratio (OR) with 95% confidence intervals (CI). Results: Twelve studies with a total of 14 364 patients met the inclusion criteria. The pooled prevalence of obesity in patients with COVID-19 was 32.0% (95% CI, 26%-38%, P < .001). The prevalence of obesity in ICU COVID-19 patients were 37.0% (95% CI, 29%-46%, P < .001). Comparing between obese and non-obese patients, the meta-analysis showed that obesity was an important risk factor associated with COVID-19 patients needed for ICU care (OR: 1.36, 95% CI 1.22-1.52, P < .001). Conclusion: Obesity was highly prevalent (32.0%) in patients with COVID-19, especially in ICU patients (37.0%), and was an important risk factor for COVID-19 patients needed for ICU care.
Collapse
Affiliation(s)
| | - Hina Naz
- Ibne Seena Medical Centre, Karachi, Pakistan
| | - Sana Shamim
- Dow University of Health Sciences, Ojha, Karachi, Pakistan
| | | | - Sohail Ahmed
- The Health Department of Sindh, Government of Sindh, Karachi, Pakistan
| | | |
Collapse
|
38
|
Delmonte OM, Oguz C, Dobbs K, Myint-Hpu K, Palterer B, Abers MS, Draper D, Truong M, Kaplan IM, Gittelman RM, Zhang Y, Rosen LB, Snow AL, Dalgard CL, Burbelo PD, Imberti L, Sottini A, Quiros-Roldan E, Castelli F, Rossi C, Brugnoni D, Biondi A, Bettini LR, D'Angio M, Bonfanti P, Anderson MV, Saracino A, Chironna M, Di Stefano M, Fiore JR, Santantonio T, Castagnoli R, Marseglia GL, Magliocco M, Bosticardo M, Pala F, Shaw E, Matthews H, Weber SE, Xirasagar S, Barnett J, Oler AJ, Dimitrova D, Bergerson JRE, McDermott DH, Rao VK, Murphy PM, Holland SM, Lisco A, Su HC, Lionakis MS, Cohen JI, Freeman AF, Snyder TM, Lack J, Notarangelo LD. Perturbations of the T-cell receptor repertoire in response to SARS-CoV-2 in immunocompetent and immunocompromised individuals. J Allergy Clin Immunol 2024; 153:1655-1667. [PMID: 38154666 PMCID: PMC11162338 DOI: 10.1016/j.jaci.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Functional T-cell responses are essential for virus clearance and long-term protection after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas certain clinical factors, such as older age and immunocompromise, are associated with worse outcome. OBJECTIVE We sought to study the breadth and magnitude of T-cell responses in patients with coronavirus disease 2019 (COVID-19) and in individuals with inborn errors of immunity (IEIs) who had received COVID-19 mRNA vaccine. METHODS Using high-throughput sequencing and bioinformatics tools to characterize the T-cell receptor β repertoire signatures in 540 individuals after SARS-CoV-2 infection, 31 IEI recipients of COVID-19 mRNA vaccine, and healthy controls, we quantified HLA class I- and class II-restricted SARS-CoV-2-specific responses and also identified several HLA allele-clonotype motif associations in patients with COVID-19, including a subcohort of anti-type 1 interferon (IFN-1)-positive patients. RESULTS Our analysis revealed that elderly patients with COVID-19 with critical disease manifested lower SARS-CoV-2 T-cell clonotype diversity as well as T-cell responses with reduced magnitude, whereas the SARS-CoV-2-specific clonotypes targeted a broad range of HLA class I- and class II-restricted epitopes across the viral proteome. The presence of anti-IFN-I antibodies was associated with certain HLA alleles. Finally, COVID-19 mRNA immunization induced an increase in the breadth of SARS-CoV-2-specific clonotypes in patients with IEIs, including those who had failed to seroconvert. CONCLUSIONS Elderly individuals have impaired capacity to develop broad and sustained T-cell responses after SARS-CoV-2 infection. Genetic factors may play a role in the production of anti-IFN-1 antibodies. COVID-19 mRNA vaccines are effective in inducing T-cell responses in patients with IEIs.
Collapse
Affiliation(s)
- Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Katherine Myint-Hpu
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael S Abers
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Deborah Draper
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Meng Truong
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | | | - Yu Zhang
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Lindsey B Rosen
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrew L Snow
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md; Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Clifton L Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Md; The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Peter D Burbelo
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Md
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Alessandra Sottini
- Section of Microbiology, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Francesco Castelli
- Department of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Camillo Rossi
- Direzione Sanitaria, ASST Spedali Civili, Brescia, Italy
| | - Duilio Brugnoni
- Laboratorio Analisi Chimico-Cliniche, ASST Spedali Civili, Brescia, Italy
| | - Andrea Biondi
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Laura Rachele Bettini
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Mariella D'Angio
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Megan V Anderson
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Annalisa Saracino
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, University of Bari, Bari, Italy
| | - Maria Chironna
- Hygiene Section, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Mariantonietta Di Stefano
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Jose Ramon Fiore
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Teresa Santantonio
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Riccardo Castagnoli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gian Luigi Marseglia
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mary Magliocco
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Elana Shaw
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Helen Matthews
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sarah E Weber
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sandhya Xirasagar
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jason Barnett
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrew J Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dimana Dimitrova
- Center for Immuno-Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Jenna R E Bergerson
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - David H McDermott
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - V Koneti Rao
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | - Justin Lack
- Integrated Data Sciences Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
39
|
Chen Y, Li H, Lin J, Su Z, Lin T. Association between (ΔPaO2/FiO2)/PEEP and in-hospital mortality in patients with COVID-19 pneumonia: A secondary analysis. PLoS One 2024; 19:e0304518. [PMID: 38820377 PMCID: PMC11142544 DOI: 10.1371/journal.pone.0304518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND The arterial pressure of oxygen (PaO2)/inspiratory fraction of oxygen (FiO2) is associated with in-hospital mortality in patients with Coronavirus Disease 2019 (COVID-19) pneumonia. ΔPaO2/FiO2 [the difference between PaO2/FiO2 after 24 h of invasive mechanical ventilation (IMV) and PaO2/FiO2 before IMV] is associated with in-hospital mortality. However, the value of PaO2 can be influenced by the end-expiratory pressure (PEEP). To the best of our knowledge, the relationship between the ratio of (ΔPaO2/FiO2)/PEEP and in-hospital mortality remains unclear. This study aimed to evaluate their association. METHODS The study was conducted in southern Peru from April 2020 to April 2021. A total of 200 patients with COVID-19 pneumonia requiring IMV were included in the present study. We analyzed the association between (ΔPaO2/FiO2)/PEEP and in-hospital mortality by Cox proportional hazards regression models. RESULTS The median (ΔPaO2/FiO2)/PEEP was 11.78 mmHg/cmH2O [interquartile range (IQR) 8.79-16.08 mmHg/cmH2O], with a range of 1 to 44.36 mmHg/cmH2O. Patients were divided equally into two groups [low group (< 11.80 mmHg/cmH2O), and high group (≥ 11.80 mmHg/cmH2O)] according to the (ΔPaO2/FiO2)/PEEP ratio. In-hospital mortality was lower in the high (ΔPaO2/FiO2)/PEEP group than in the low (ΔPaO2/FiO2)/PEEP group [18 (13%) vs. 38 (38%)]; hazard ratio (HR), 0.33 [95% confidence intervals (CI), 0.17-0.61, P < 0.001], adjusted HR, 0.32 (95% CI, 0.11-0.94, P = 0.038). The finding that the high (ΔPaO2/FiO2)/PEEP group exhibited a lower risk of in-hospital mortality compared to the low (ΔPaO2/FiO2)/PEEP group was consistent with the results from the sensitivity analysis. After adjusting for confounding variables, we found that each unit increase in (ΔPaO2/FiO2)/PEEP was associated with a 12% reduction in the risk of in-hospital mortality (HR, 0.88, 95%CI, 0.80-0.97, P = 0.013). CONCLUSIONS The (ΔPaO2/FiO2)/PEEP ratio was associated with in-hospital mortality in patients with COVID-19 pneumonia. (ΔPaO2/FiO2)/PEEP might be a marker of disease severity in COVID-19 patients.
Collapse
Affiliation(s)
- Youli Chen
- Intensive Care Unit, Fujian Medical University Affiliated First Quanzhou Hospital, Quanzhou, Fujian, PR China
| | - Huangen Li
- Intensive Care Unit, Fujian Medical University Affiliated First Quanzhou Hospital, Quanzhou, Fujian, PR China
| | - Jinhuang Lin
- Intensive Care Unit, Fujian Medical University Affiliated First Quanzhou Hospital, Quanzhou, Fujian, PR China
| | - Zhiwei Su
- Intensive Care Unit, Fujian Medical University Affiliated First Quanzhou Hospital, Quanzhou, Fujian, PR China
| | - Tianlai Lin
- Intensive Care Unit, Fujian Medical University Affiliated First Quanzhou Hospital, Quanzhou, Fujian, PR China
| |
Collapse
|
40
|
Saconato M, Maselli-Schoueri JH, Malaque CMS, Marcusso RM, de Oliveira ACP, Batista LAN, Ultramari G, Lindoso JAL, Gonçalves MIR, Sztajnbok J. Postorotracheal intubation dysphagia in patients with COVID-19: A retrospective study. SAO PAULO MED J 2024; 142:e2022608. [PMID: 38808794 PMCID: PMC11126317 DOI: 10.1590/1516-3180.2022.0608.r3.14032024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/09/2023] [Accepted: 03/14/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The cause of oropharyngeal dysphagia in patients with coronavirus disease (COVID-19) can be multifactorial and may underly limitations in swallowing rehabilitation. OBJECTIVE Analyze the factors related to dysphagia in patients with COVID-19 immediately after orotracheal extubation and the factors that influence swallowing rehabilitation. DESIGN AND SETTING A retrospective study. METHODS The presence of dysphagia was evaluated using the American Speech-Language Hearing Association National Outcome Measurement System (ASHA NOMS) scale and variables that influenced swallowing rehabilitation in 140 adult patients who required invasive mechanical ventilation for >48 h. RESULTS In total, 46.43% of the patients scored 1 or 2 on the ASHA NOMS (severe dysphagia) and 39.29% scored 4 (single consistency delivered orally) or 5 (exclusive oral diet with adaptations). Both the length of mechanical ventilation and the presence of neurological disorders were associated with lower ASHA NOMS scores (odds ratio [OR]: 0.80, 95% confidence interval [CI]: 0.74-0.87 P < 0.05; and OR: 0.13, 95% CI: 0.61-0.29; P < 0.05, respectively). Age and the presence of tracheostomy were negatively associated with speech rehabilitation (OR: 0.92; 95% CI: 0.87--0.96; OR: 0.24; 95% CI: 0.80--0.75), and acute post-COVID-19 kidney injury requiring dialysis and lower scores on the ASHA NOMS were associated with longer time for speech therapy outcomes (β: 1.62, 95% CI, 0.70-3.17, P < 0.001; β: -1.24, 95% CI: -1.55--0.92; P < 0.001). CONCLUSION Prolonged orotracheal intubation and post-COVID-19 neurological alterations increase the probability of dysphagia immediately after extubation. Increased age and tracheostomy limited rehabilitation.
Collapse
Affiliation(s)
- Mariana Saconato
- PhD. Speech therapist, Technical manager of the Speech Therapy team, Instituto de Infectologia Emílio Ribas (IIER), São Paulo (SP), Brazil
| | | | - Ceila Maria Sant’Ana Malaque
- PhD. Physician, Intensive Care Unit Physician, Instituto de Infectologia Emílio Ribas (IIER), São Paulo (SP), Brazil
| | - Rosa Maria Marcusso
- MSc. Statistician, Instituto de Infectologia Emílio Ribas (IIER), São Paulo (SP), Brazil
| | | | | | - Graziela Ultramari
- MSc. Physiotherapist, Head of the Diagnostic and Therapeutic Support Department, Instituto de Infectologia Emílio Ribas (IIER), São Paulo (SP), Brazil
| | - José Angelo Lauletta Lindoso
- PhD. Physician, Director of the Diagnostic and Therapeutic Support Department, Instituto de Infectologia Emílio Ribas (IIER), São Paulo (SP), Brazil
| | - Maria Inês Rebelo Gonçalves
- PhD. Speech therapist and Professor, Department of Speech Therapy, Universidade Federal de São Paulo (UNIFESP), São Paulo (SP), Brazil
| | - Jaques Sztajnbok
- MD. Physician, Head of the Intensive Care Unit, Instituto de Infectologia Emílio Ribas (IIER), São Paulo (SP), Brazil
| |
Collapse
|
41
|
Oh AR, Kang ES, Park J, Lee SM, Jeong M, Lee JH. Does coronavirus disease 2019 history alone increase the risk of postoperative pulmonary complications after surgery? Prospective observational study using serology assessment. PLoS One 2024; 19:e0300782. [PMID: 38771760 PMCID: PMC11108156 DOI: 10.1371/journal.pone.0300782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/05/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Concern exists about the increasing risk of postoperative pulmonary complications in patients with a history of coronavirus disease 2019 (COVID-19). OBJECTIVE We conducted a prospective observational study that compared the incidence of postoperative pulmonary complications in patients with and without a history of COVID-19. METHODS From August 2022 to November 2022, 244 adult patients undergoing major non-cardiac surgery were enrolled and allocated either to history or no history of COVID-19 groups. For patients without a history of confirming COVID-19 diagnosis, we tested immunoglobulin G to nucleocapsid antigen of SARS-CoV-2 for serology assessment to identify undetected infection. We compared the incidence of postoperative pulmonary complications, defined as a composite of atelectasis, pleural effusion, pulmonary edema, pneumonia, aspiration pneumonitis, and the need for additional oxygen therapy according to a COVID-19 history. RESULTS After excluding 44 patients without a COVID-19 history who were detected as seropositive, 200 patients were finally enrolled in this study, 100 in each group. All subjects with a COVID-19 history experienced no or mild symptoms during infection. The risk of postoperative pulmonary complications was not significantly different between the groups according to the history of COVID-19 (24.0% vs. 26.0%; odds ratio, 0.99; 95% confidence interval, 0.71-1.37; P-value, 0.92). The incidence of postoperative pulmonary complications was also similar (27.3%) in excluded patients owing to being seropositive. CONCLUSION Our study showed patients with a history of no or mild symptomatic COVID-19 did not show an increased risk of PPCs compared to those without a COVID-19 history. Additional precautions may not be needed to prevent PPCs in those patients.
Collapse
Affiliation(s)
- Ah Ran Oh
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun-Suk Kang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jungchan Park
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sangmin Maria Lee
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mijeong Jeong
- Research Institute for Future Medicine, R&D Management & Supporting Team, Republic of Korea
| | - Jong-Hwan Lee
- Department of Anesthesiology and Pain Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
42
|
Aung MO, Venkatachalam I, Sim JX, Wee LE, Aung MK, Yang Y, Conceicao EP, Arora S, Lee MA, Sia CH, Tan KB, Ling ML. Prediction model to identify infectious COVID-19 patients in the emergency department. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2024; 4:e88. [PMID: 38774116 PMCID: PMC11106730 DOI: 10.1017/ash.2024.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024]
Abstract
Background Real-time reverse-transcriptase polymerase chain reaction (RT-PCR) has been the gold standard for diagnosing coronavirus disease 2019 (COVID-19) but has a lag time for the results. An effective prediction algorithm for infectious COVID-19, utilized at the emergency department (ED), may reduce the risk of healthcare-associated COVID-19. Objective To develop a prototypic prediction model for infectious COVID-19 at the time of presentation to the ED. Material and methods Retrospective cohort study of all adult patients admitted to Singapore General Hospital (SGH) through ED between March 15, 2020, and December 31, 2022, with admission of COVID-19 RT-PCR results. Two prediction models were developed and evaluated using area under the curve (AUC) of receiver operating characteristics (ROC) to identify infectious COVID-19 patients (cycle threshold (Ct) of <25). Results Total of 78,687 patients were admitted to SGH through ED during study period. 6,132 of them tested severe acute respiratory coronavirus 2 positive on RT-PCR. Nearly 70% (4,226 of 6,132) of the patients had infectious COVID-19 (Ct<25). Model that included demographics, clinical history, symptom and laboratory variables had AUROC of 0.85 with sensitivity and specificity of 80.0% & 72.1% respectively. When antigen rapid test results at ED were available and added to the model for a subset of the study population, AUROC reached 0.97 with sensitivity and specificity of 95.0% and 92.8% respectively. Both models maintained respective sensitivity and specificity results when applied to validation data. Conclusion Clinical predictive models based on available information at ED can be utilized for identification of infectious COVID-19 patients and may enhance infection prevention efforts.
Collapse
Affiliation(s)
- Myat Oo Aung
- Infection Prevention and Epidemiology Department, Singapore General Hospital, Singapore, Singapore
| | - Indumathi Venkatachalam
- Infection Prevention and Epidemiology Department, Singapore General Hospital, Singapore, Singapore
- Department of Infectious Disease, Singapore General Hospital, Singapore, Singapore
| | - Jean X.Y. Sim
- Infection Prevention and Epidemiology Department, Singapore General Hospital, Singapore, Singapore
- Department of Infectious Disease, Singapore General Hospital, Singapore, Singapore
| | - Liang En Wee
- Department of Infectious Disease, Singapore General Hospital, Singapore, Singapore
| | - May K. Aung
- Infection Prevention and Epidemiology Department, Singapore General Hospital, Singapore, Singapore
| | - Yong Yang
- Infection Prevention and Epidemiology Department, Singapore General Hospital, Singapore, Singapore
| | - Edwin P. Conceicao
- Infection Prevention and Epidemiology Department, Singapore General Hospital, Singapore, Singapore
| | - Shalvi Arora
- Infection Prevention and Epidemiology Department, Singapore General Hospital, Singapore, Singapore
| | - Marcus A.B. Lee
- Emergency Department, Singapore General Hospital, Singapore, Singapore
| | - Chang H. Sia
- Emergency Department, Singapore General Hospital, Singapore, Singapore
| | - Kenneth B.K. Tan
- Emergency Department, Singapore General Hospital, Singapore, Singapore
| | - Moi Lin Ling
- Infection Prevention and Epidemiology Department, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
43
|
Singh T, Rao Padubidri J, Shetty PH, Antony Manoj M, Mary T, Thejaswi Pallempati B. The top 50 most-cited articles about COVID-19 and the complications of COVID-19: A bibliometric analysis. F1000Res 2024; 13:105. [PMID: 39149509 PMCID: PMC11325134 DOI: 10.12688/f1000research.145713.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 08/17/2024] Open
Abstract
Background This bibliometric analysis examines the top 50 most-cited articles on COVID-19 complications, offering insights into the multifaceted impact of the virus. Since its emergence in Wuhan in December 2019, COVID-19 has evolved into a global health crisis, with over 770 million confirmed cases and 6.9 million deaths as of September 2023. Initially recognized as a respiratory illness causing pneumonia and ARDS, its diverse complications extend to cardiovascular, gastrointestinal, renal, hematological, neurological, endocrinological, ophthalmological, hepatobiliary, and dermatological systems. Methods Identifying the top 50 articles from a pool of 5940 in Scopus, the analysis spans November 2019 to July 2021, employing terms related to COVID-19 and complications. Rigorous review criteria excluded non-relevant studies, basic science research, and animal models. The authors independently reviewed articles, considering factors like title, citations, publication year, journal, impact factor, authors, study details, and patient demographics. Results The focus is primarily on 2020 publications (96%), with all articles being open access. Leading journals include The Lancet, NEJM, and JAMA, with prominent contributions from Internal Medicine (46.9%) and Pulmonary Medicine (14.5%). China played a major role (34.9%), followed by France and Belgium. Clinical features were the primary study topic (68%), often utilizing retrospective designs (24%). Among 22,477 patients analyzed, 54.8% were male, with the most common age group being 26-65 years (63.2%). Complications of COVID-19 affected 13.9% of patients, with a recovery rate of 57.8%. Conclusion Analyzing these top-cited articles offers clinicians and researchers a comprehensive, timely understanding of influential COVID-19 literature. This approach uncovers attributes contributing to high citations and provides authors with valuable insights for crafting impactful research. As a strategic tool, this analysis facilitates staying updated and making meaningful contributions to the dynamic field of COVID-19 research.
Collapse
Affiliation(s)
- Tanya Singh
- Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Jagadish Rao Padubidri
- Department of Forensic Medicine and Toxicology, Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Pavanchand H Shetty
- Department of Forensic Medicine and Toxicology, Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Matthew Antony Manoj
- Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Therese Mary
- Kasturba Medical College Mangalore, Manipal Academy of Higher Education, Manipal, India
| | | |
Collapse
|
44
|
Berry GJ, Jhaveri TA, Larkin PMK, Mostafa H, Babady NE. ADLM Guidance Document on Laboratory Diagnosis of Respiratory Viruses. J Appl Lab Med 2024; 9:599-628. [PMID: 38695489 DOI: 10.1093/jalm/jfae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 06/06/2024]
Abstract
Respiratory viral infections are among the most frequent infections experienced worldwide. The COVID-19 pandemic has highlighted the need for testing and currently several tests are available for the detection of a wide range of viruses. These tests vary widely in terms of the number of viral pathogens included, viral markers targeted, regulatory status, and turnaround time to results, as well as their analytical and clinical performance. Given these many variables, selection and interpretation of testing requires thoughtful consideration. The current guidance document is the authors' expert opinion based on the preponderance of available evidence to address key questions related to best practices for laboratory diagnosis of respiratory viral infections including who to test, when to test, and what tests to use. An algorithm is proposed to help laboratories decide on the most appropriate tests to use for the diagnosis of respiratory viral infections.
Collapse
Affiliation(s)
- Gregory J Berry
- Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian-Columbia University Irving Medical Center, New York, NY, United States
| | - Tulip A Jhaveri
- Department of Internal Medicine, Division of Infectious Diseases, University of Mississippi Medical Center, Jackson, MS, United States
| | - Paige M K Larkin
- University of Chicago Pritzker School of Medicine, NorthShore University Health System, Chicago, IL, United States
| | - Heba Mostafa
- Johns Hopkins School of Medicine, Department of Pathology, Baltimore, MD, United States
| | - N Esther Babady
- Clinical Microbiology and Infectious Disease Services, Department of Pathology and Laboratory Medicine and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
45
|
Chen Y, Liu J, Shao S, Song Z, Ma Y, Tuo Y, Fang L, Xu Y, Xu B, Gu W, Cao X, Chen J, Yang Y, Wang P, Zhang J, Xu Y, Yu D, Hou P, Meng K, Li Z, Liu G, Qu X, Ji L, Yang R, Zhang L. Characteristics and outcomes of COVID-19 in Chinese immune thrombocytopenia patients: A prospective cohort study. Br J Haematol 2024; 204:1207-1218. [PMID: 37967471 DOI: 10.1111/bjh.19198] [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: 09/04/2023] [Revised: 10/09/2023] [Accepted: 10/30/2023] [Indexed: 11/17/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has a significant impact on the immune system. This is the first and largest study on pre-existing immune thrombocytopenia (ITP) patients infected with COVID-19 in China. We prospectively collected ITP patients infected with COVID-19 enrolled in the National Longitudinal Cohort of Hematological Diseases (NICHE, NCT04645199) and followed up for at least 1 month after infection. One thousand and one hundred forty-eight pre-existing ITP patients were included. Two hundred and twelve (18.5%) patients showed a decrease in the platelet (PLT) count after infection. Forty-seven (4.1%) patients were diagnosed with pneumonia. Risk factors for a decrease in the PLT count included baseline PLT count <50 × 109/L (OR, 1.76; 95% CI, 1.25-2.46; p = 0.001), maintenance therapy including thrombopoietin receptor agonists (TPO-RAs) (OR, 2.27; 95% CI, 1.60-3.21; p < 0.001) and previous splenectomy (OR, 1.98; 95% CI, 1.09-3.61; p = 0.03). Risk factors for pneumonia included age ≥40 years (OR, 2.45; 95% CI, 1.12-5.33; p = 0.02), ≥2 comorbidities (OR, 3.47; 95% CI, 1.63-7.64; p = 0.001), maintenance therapy including TPO-RAs (OR, 2.14; 95% CI, 1.17-3.91; p = 0.01) and immunosuppressants (OR, 3.05; 95% CI, 1.17-7.91; p = 0.02). In this cohort study, we described the characteristics of pre-existing ITP patients infected with COVID-19 and identified several factors associated with poor outcomes.
Collapse
Affiliation(s)
- Yunfei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jiaying Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuai Shao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhen Song
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yueshen Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuanyuan Tuo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Department of Pediatric Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lijun Fang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yanmei Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Bingqi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenjing Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xuan Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jia Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Panjing Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jing Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dandan Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Pengxiao Hou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ke Meng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhirong Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Guanyu Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xinmiao Qu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lulu Ji
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
46
|
Li N, Zhang L, Gao Y, Lai Q, Tang Y, Du X, Chen P, Yue C, Zhao M, Yu K, Kang K. Changes in clinical parameters and inflammatory markers after blood culture collection facilitate early identification of positive culture in adult patients with COVID-19 and clinically suspected bloodstream infection. J Int Med Res 2024; 52:3000605241238134. [PMID: 38630560 PMCID: PMC11025435 DOI: 10.1177/03000605241238134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 02/22/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE We explored whether changes in clinical parameters and inflammatory markers can facilitate early identification of positive blood culture in adult patients with COVID-19 and clinically suspected bloodstream infection (BSI). METHODS This single-center retrospective study enrolled 20 adult patients with COVID-19 admitted to the intensive care unit who underwent blood culture for clinically suspected BSI (February 2020-November 2021). We divided patients into positive (Pos) and negative blood culture groups. Clinical parameters and inflammatory markers were obtained from medical records between blood culture collection and the first positive or negative result and compared between groups on different days. RESULTS Patients in the positive culture group had significantly older age and higher D-dimer, immunoglobulin 6 (IL-6), and Sequential Organ Failure Assessment score as well as lower albumin (ALB). The area under the receiver operating characteristic curve (AUC) was 0.865 for IL-6, D-dimer and ALB on the first day after blood culture collection; the AUC was 0.979 for IL-6, IL-10, D-dimer, and C-reactive protein on the second day after blood culture collection. CONCLUSION Changes in clinical parameters and inflammatory markers after blood culture collection may facilitate early identification of positive culture in adult patients with COVID-19 and clinically suspected BSI.
Collapse
Affiliation(s)
- Nana Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Long Zhang
- Department of Critical Care Medicine, The Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Yang Gao
- Department of Critical Care Medicine, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Institute of Critical Care Medicine, The Sino Russian Medical Research Center of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qiqi Lai
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yujia Tang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xue Du
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Pengfei Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Chuangshi Yue
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Institute of Critical Care Medicine, The Sino Russian Medical Research Center of Harbin Medical University, Harbin, Heilongjiang Province, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang Province, China
- Key Laboratory of Cell Transplantation, National Health Commission, Harbin, Heilongjiang Province, China
| | - Kai Kang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
47
|
Bobot M, Hak JF, Casolla B, Dehondt JD, Burtey S, Doche E, Suissa L. Acute and Chronic Kidney Dysfunction and Prognosis following Thrombectomy for Ischemic Stroke. Am J Nephrol 2024; 55:287-297. [PMID: 38499002 DOI: 10.1159/000536493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/10/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Patients with chronic kidney disease (CKD) have an increased risk of stroke, and CKD seems associated with worse outcome after a stroke. The main objective of our study RISOTTO was to evaluate the influence of CKD and acute kidney injury (AKI) on the clinical outcome and mortality of ischemic stroke patients after thrombolysis and/or thrombectomy. METHODS This multicenter cohort study included patients in the acute phase of ischemic stroke due to large artery occlusion managed by thrombectomy. Functional outcome at 3 months was assessed by the modified Rankin Scale (mRS). RESULTS 280 patients were included in the analysis. Fifty-nine patients (22.6%) had CKD. At 3 months, CKD was associated with similar functional prognosis (mRS 3-6: 50.0% vs. 41.7%, p = 0.262) but higher mortality (24.2% versus 9.5%, p = 0.004). In univariate analysis, patients with CKD had a higher burden of white matter hyperintensities (Fazekas score: 1.7 ± 0.8 vs. 1.0 ± 0.8, p = 0.002), lower initial infarct volume with equivalent severity, and lower recanalization success (86.4% vs. 97.0%, p = 0.008) compared to non-CKD patients. Forty-seven patients (20.0%) developed AKI. AKI was associated with poorer 3-month functional outcome (mRS 3-6: 63.8% vs. 49.0%, p = 0.002) and mortality (23.4% versus 7.7%, p = 0.002). In multivariate analysis, AKI appeared as an independent risk factor for poor functional outcome (mRS 3-6: adjOR 2.79 [1.11-7.02], p = 0.029) and mortality (adjOR 2.52 [1.03-6.18], p = 0.043) at 3 months, while CKD was not independently associated with 3-month mortality and poor neurological outcome. CONCLUSIONS AKI is independently associated with poorer functional outcome and increased mortality at 3 months. CKD was not an independent risk factor for 3-month mortality or poor functional prognosis.
Collapse
Affiliation(s)
- Mickaël Bobot
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, AP-HM, Marseille, France
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- CERIMED, Aix Marseille Univ, Marseille, France
| | - Jean-François Hak
- CERIMED, Aix Marseille Univ, Marseille, France
- Neuroradiology Department, CHU Timone, AP-HM, Marseille, France
- LIIE, CERIMED, Aix-Marseille Université, Marseille, France
| | - Barbara Casolla
- Stroke Unit, UR2CA-URRIS Neurology, Nice Cote d'Azur University, Nice, France
| | | | - Stéphane Burtey
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, AP-HM, Marseille, France
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Emilie Doche
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Stroke Center, Hôpital de la Timone, AP-HM, Marseille, France
| | - Laurent Suissa
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Stroke Center, Hôpital de la Timone, AP-HM, Marseille, France
| |
Collapse
|
48
|
Jahanshahi F, Jazayeri SB, Eraghi MM, Reis LO, Hamidikia M, Amiri S, Aghamir SMK. A narrative review on adverse drug reactions of COVID-19 treatments on the kidney. Open Med (Wars) 2024; 19:20230867. [PMID: 38584847 PMCID: PMC10996932 DOI: 10.1515/med-2023-0867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 11/01/2022] [Accepted: 11/18/2023] [Indexed: 04/09/2024] Open
Abstract
Studies showed that the respiratory is not the only system affected by coronavirus 2, while cardiovascular, digestive, and nervous systems, as well as essential organs such as the kidneys, can be affected by this virus. In this review, we have studied the epidemiology, clinical, and laboratory findings on COVID-19 infection renal involvement, mortality, physiopathology, remaining renal sequels after recovery, underlying renal disease, and renal injury due to its treatment. Also, protective measures for kidney injury are explained in three levels. Evidence of viral particles and genome in the urine and renal tubular cells and signs of damage such as microangiopathy, hypercoagulopathy, and fibrosis are found in COVID-19 patients. The result of this study showed, in hospitalized COVID-19 patients, that the rate of acute kidney injury (AKI) was up to 46%, with a mortality ranging from 11 to 96%. A considerable proportion of patients with AKI would remain on renal replacement therapy. Proteinuria and hematuria are observed in 87 and 75% patients, and increased Cr and glomerular filtration rate (GFR) <60 ml/min per 1.73 m2 are observed in 29.6 and 35.3% of the patients, respectively. Remedsivir is considered to have adverse effects on GFR. COVID-19 patients need special attention to prevent AKI. Those with underlying chronic kidney disease or AKI need proper and explicit evaluation and treatment to improve their prognosis and decrease mortality, which should not be limited to the hospitalization period.
Collapse
Affiliation(s)
- Fatemeh Jahanshahi
- Research Committee Member, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Urology Research Center, Tehran University of Medical Sciences, Sina Hospital, Tehran, Iran
| | - Seyed Behnam Jazayeri
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mirahmadi Eraghi
- Urology Research Center, Tehran University of Medical Sciences, Sina Hospital, Tehran, Iran
- School of Medicine, Qeshm International Branch, Islamic Azad University, Qeshm, Iran
| | - Leonardo Oliveira Reis
- UroScience and Department of Surgery (Urology), School of Medical Sciences, University of Campinas, Unicamp, and Pontifical Catholic University of Campinas, PUC-Campinas, Campinas, São Paulo, Brazil
| | - Mahtab Hamidikia
- Research Committee Member, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- Rasool Akram Medical Complex, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
49
|
Lin C, Li M, Lin Y, Zhang Y, Xu H, Chen B, Yan X, Xu Y. Impact of plasma Epstein-Barr virus DNA in posttreatment nasopharyngeal carcinoma patients after SARS-CoV-2 infection. Infect Agent Cancer 2024; 19:8. [PMID: 38486290 PMCID: PMC10938826 DOI: 10.1186/s13027-024-00570-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is prevalent in southern China. EBV DNA is the most useful biomarker in NPC. However, the value of EBV DNA in posttreatment NPC patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains unclear. METHODS Sixty-four eligible NPC patients were enrolled between December 2022 and February 2023. Patients who met the following criteria were included: had non-metastatic NPC, completed radical treatment, were first firstly infected with SARS-CoV-2 and their EBV DNA changed from undetectable to detectable. RESULTS At the end of follow-up, 81.25% (52/64) of patients were confirmed not to relapse with undetectable EBV DNA (no-relapse). In addition, 18.75% (12/64) of patients experienced relapse with consistent detection of EBV DNA (yes-relapse). For all 64 patients, the average time from diagnosis of coronavirus disease 2019 (COVID-19) to detection of detectable EBV DNA was 35.41 days (2 to 139 days). For 52 no-relapse patients, the average time from EBV DNA changing from detectable to undetectable was 63.12 days (6 to 147 days). The levels of EBV DNA were greater in yes-relapse patients than that in no-relapse patients, and the average of EBV DNA levels were 1216 copies/ml and 53.18 copies/ml, respectively. Using 62.3 copies/mL as the threshold, the area under the curve for EBV DNA was 0.88 for distinguishing yes-relapse patients from no-relapse patients. The sensitivity and specificity were 81.97% (95% CI 0.71-0.95) and 86.67% (95% CI 0.70-0.95), respectively. CONCLUSION For NPC patients infected with SARS-CoV-2, EBV DNA alone is insufficient for monitoring relapse after radical therapy. Long-term follow-up and underlying mechanistic investigations of EBV DNA changes are urgently needed.
Collapse
Affiliation(s)
- Cheng Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
- Interdisciplinary College of Medicine and Engineering, Fuzhou University, Fuzhou, Fujian, China
| | - Meifang Li
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Yingying Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Yu Zhang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Hanchuan Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Bijuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Xia Yan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Yun Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China.
| |
Collapse
|
50
|
Butler MJ, Chiuzan C, Ahn H, Gao M, D’Angelo S, Yeh J, Davidson K. Before and after COVID-19: Changes in symptoms and diagnoses in 13,033 adults. PLoS One 2024; 19:e0286371. [PMID: 38457409 PMCID: PMC10923490 DOI: 10.1371/journal.pone.0286371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/15/2023] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Most patients with COVID-19 report experiencing one or more symptoms after acute infection subsides, known as post-acute sequelae of SARS-CoV-2 infection (PASC). Though research has examined PASC after acute COVID-19, few studies have examined PASC over a longer follow-up duration or accounted for rates of symptoms and diagnoses before COVID-19 infection, and included those not actively seeking treatment for PASC. To determine what symptoms and diagnoses are occurring at higher rates after acute COVID-19 infection from a more inclusive sample, we extracted electronic hospital records (EHR) data from 13,033 adults with previously known diagnoses and symptoms. METHODS The sample was comprised of patients who had a positive PCR test for SARS-CoV-2 between March 1, 2020, and December 31, 2020, and follow-up was conducted through November 29, 2021. All patients in the sample had medical appointments ≥4 weeks before and ≥4 weeks after their positive PCR test. At these appointments, all ICD-10 codes recorded in the EHR were classified into 21 categories based on the literature and expert review. Conditional logistic regression models were used to quantify the odds of these symptoms and diagnostic categories following COVID-19 infection relative to visits occurring before infection. The sample was comprised of 28.0% adults over 65 and was 57.0% female. After the positive PCR test, the most recorded diagnoses and symptoms were dyspnea and respiratory failure, myositis, musculoskeletal pain/stiffness, anxiety, and depression. RESULTS Results from regression analyses showed increased odds of diagnosis for 15 of the 21 categories following positive PCR. Relative to pre-COVID, the diagnoses and symptoms with the greatest odds after a positive PCR test were loss of smell or taste [OR (95% CI) = 6.20 (3.18-12.09)], pulmonary fibrosis [3.50 (1.59-7.68)], and dyspnea/respiratory failure [2.14 (1.92-2.40)]. Stratification of these analyses by age, gender, race, and ethnicity showed similar results. CONCLUSION The increased symptoms and diagnoses detected in the current study match prior analyses of PASC diagnosis and treatment-seeking patients. The current research expands upon the literature by showing that these symptoms are more frequently detected following acute COVID-19 than before COVID-19. Further, our analyses provide a broad snapshot of the population as we were able to describe PASC among all patients who tested positive for COVID-19.
Collapse
Affiliation(s)
- Mark J. Butler
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, New York, NY, United States of America
| | - Codruta Chiuzan
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, New York, NY, United States of America
| | - Heejoon Ahn
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, New York, NY, United States of America
| | - Michael Gao
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, New York, NY, United States of America
| | - Stefani D’Angelo
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, New York, NY, United States of America
| | - Jackson Yeh
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, New York, NY, United States of America
| | - Karina Davidson
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, New York, NY, United States of America
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States of America
| |
Collapse
|