1
|
Carpallo-Porcar B, Jiménez-Sánchez C, Calvo S, Irún P, Kolesnyk-Sumskaya E, Aller-Blanco AI, Beamonte EDC. ARACOV-02. Specialized nutritional intervention and telerehabilitation in patients with long COVID: Protocol of a randomized controlled trial. PLoS One 2025; 20:e0321811. [PMID: 40299883 PMCID: PMC12040102 DOI: 10.1371/journal.pone.0321811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/17/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND It is estimated that at least 10% of the population infected with SARS-CoV-2 develop Post COVID Condition, which is characterized by a diverse array of symptoms including dyspnea, fatigue, anxiety, depression, and deterioration in quality of life. The SARS-CoV-2 virus can trigger an excessive immune response, characterized by the release of pro-inflammatory cytokines including IL-6, IL-1, TNFα and reactive oxygen species. Specialized Pro Resolving Mediators (SPMs) (17-HAD, 14-HAD and 18_HEPE) that could be useful in Post COVID Condition modulating the inflammatory response. The objective is to determine the change in quality of life, inflammatory profile, functional capacity and emotional variables in a group taking a nutritional supplement (SPMs) plus a telerehabilitation programme. METHODS ARACOV-2 study is a double-blind, parallel-group, randomized control trial with two parallel interventions: Nutritional supplement and telerehabilitation vs placebo supplement and telerehabilitation. The primary endpoint will be quality of life (EQ-5L-5D). The intervention will last 12 weeks with a daily intake of omega-3 or placebo and a daily supervised rehabilitation programme using telerehabilitation. DISCUSSION This study suggests that SPMs supplementation combined with telerehabilitation may improve inflammation and symptoms like fatigue in Post COVID Condition patients. Both interventions have anti-inflammatory potential, and their combined use could enhance physical and mental health outcomes. This approach offers a promising strategy for managing Post COVID Condition symptoms. TRIAL REGISTRATION ClinicalTrials.gov NCT06063031.
Collapse
Affiliation(s)
- Beatriz Carpallo-Porcar
- Department of Physical Therapy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain
| | - Carolina Jiménez-Sánchez
- Department of Physical Therapy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, Spain
| | - Sandra Calvo
- IIS Aragón, Zaragoza, Spain
- Department of Physiatry and Nursing, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
| | - Pilar Irún
- IIS Aragón, Zaragoza, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | | | | | | |
Collapse
|
2
|
de Melo BP, da Silva JAM, Rodrigues MA, Palmeira JDF, Amato AA, Argañaraz GA, Argañaraz ER. SARS-CoV-2 Spike Protein and Long COVID-Part 2: Understanding the Impact of Spike Protein and Cellular Receptor Interactions on the Pathophysiology of Long COVID Syndrome. Viruses 2025; 17:619. [PMID: 40431631 PMCID: PMC12115913 DOI: 10.3390/v17050619] [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: 02/28/2025] [Revised: 04/06/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
SARS-CoV-2 infection has had a significant impact on global health through both acute illness, referred to as coronavirus disease 2019 (COVID-19), and chronic conditions (long COVID or post-acute sequelae of COVID-19, PASC). Despite substantial advancements in preventing severe COVID-19 cases through vaccination, the rise in the prevalence of long COVID syndrome and a notable degree of genomic mutation, primarily in the S protein, underscores the necessity for a deeper understanding of the underlying pathophysiological mechanisms related to the S protein of SARS-CoV-2. In this review, the latest part of this series, we investigate the potential pathophysiological molecular mechanisms triggered by the interaction between the spike protein and cellular receptors. Therefore, this review aims to provide a differential and focused view on the mechanisms potentially activated by the binding of the spike protein to canonical and non-canonical receptors for SARS-CoV-2, together with their possible interactions and effects on the pathogenesis of long COVID.
Collapse
Affiliation(s)
- Bruno Pereira de Melo
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Jhéssica Adriane Mello da Silva
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Mariana Alves Rodrigues
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Julys da Fonseca Palmeira
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Angélica Amorim Amato
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Gustavo Adolfo Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Enrique Roberto Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| |
Collapse
|
3
|
Renner K, Stauffenberg F, Paulus M, Neumayer S, Winter-Köhler F, Buchtler S, Schmalenberger D, Blaas S, Mohr A, Pfeifer M, Malfertheiner MV, Loew T, Sester M, Bals R, Peterhoff D, Schmidt B, Mack M. Hyper-reactivity of CD8 + T cells and high expression of IL-3 correlates with occurrence and severity of Long-COVID. Clin Immunol 2025; 277:110502. [PMID: 40287027 DOI: 10.1016/j.clim.2025.110502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Following SARS-CoV-2 infection, some individuals develop Long-COVID-syndrome lasting for more than 3 months. We analyzed blood samples from patients with Long-COVID, controls without persistent symptoms following SARS-CoV-2-infection and non-infected donors without a history of infection. Long-COVID patients showed clear signs of T cell hyper-activation predominantly in the CD8+ T cell subset with a 4-fold higher expression of CD25 and 2-fold more effector-memory T cells. Following polyclonal T cell stimulation, we found a 2-fold stronger upregulation of CD25 and a 7-fold higher release of IL-3 in Long-COVID. Intracellular staining revealed 5-fold more IL-3-expressing CD8+ T cells in Long-COVID, while GM-CSF, IFN-γ and IL-2 were much less upregulated. These changes correlated with the severity of Long-COVID and persisted for up to 18 months after infection. Our data reveal a pronounced and long-lasting CD8+ T cell hyper-activation and hyper-reactivity in Long-COVID and speak for a trial of T cell-immunosuppression in patients with Long-COVID.
Collapse
Affiliation(s)
- Kerstin Renner
- University Hospital Regensburg, Department of Nephrology, 93042 Regensburg, Germany
| | - Franz Stauffenberg
- University Hospital Regensburg, Department of Nephrology, 93042 Regensburg, Germany
| | - Moritz Paulus
- University Hospital Regensburg, Department of Nephrology, 93042 Regensburg, Germany
| | - Sophia Neumayer
- University Hospital Regensburg, Department of Nephrology, 93042 Regensburg, Germany
| | | | - Simone Buchtler
- University Hospital Regensburg, Department of Nephrology, 93042 Regensburg, Germany
| | | | - Stefan Blaas
- Klinik Donaustauf, Pneumologie, 93093 Donaustauf, Germany
| | - Arno Mohr
- Klinik Donaustauf, Pneumologie, 93093 Donaustauf, Germany
| | | | | | - Thomas Loew
- Klinik Donaustauf, Psychosomatische Medizin und Psychotherapie, 93093 Donaustauf, Germany
| | - Martina Sester
- Department of Transplant and Infection Immunology, Saarland University, 66421 Homburg/Saar, Germany; Center for Gender-specific Biology and Medicine (CGBM), Saarland University, 66421 Homburg, Germany
| | - Robert Bals
- Universität des Saarlandes, Klinik für Innere Medizin V, 66421 Homburg/Saar, Germany
| | - David Peterhoff
- University of Regensburg, Institute of Clinical Microbiology and Hygiene, 93053 Regensburg, Germany
| | - Barbara Schmidt
- University of Regensburg, Institute of Clinical Microbiology and Hygiene, 93053 Regensburg, Germany
| | - Matthias Mack
- University Hospital Regensburg, Department of Nephrology, 93042 Regensburg, Germany; Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany.
| |
Collapse
|
4
|
Gysan MR, Lehmann A, Bernitzky D, Zech A, Brugger J, Prosch H, Idzko M, Gompelmann D. Immunophenotyping of bronchoalveolar lavage and functional impairment in post-COVID syndrome : Insights from a prospective cohort trial. Wien Klin Wochenschr 2025:10.1007/s00508-025-02531-9. [PMID: 40263177 DOI: 10.1007/s00508-025-02531-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE Following recovery from COVID-19, there is evidence for pulmonary sequelae and functional impairment. Data regarding the immunopathological mechanisms are limited. This study aimed to investigate the relationship between bronchoalveolar lavage fluid (BALF) cellularity, lung function impairment and high-resolution computed tomography (HRCT) changes in post-COVID syndrome patients. METHODS Patients with post-COVID syndrome were enrolled in this Austrian single-center prospective observational cohort study. All patients underwent a pulmonary function test (PFT) and chest HRCT. Those with pathological HRCT findings underwent bronchoscopy with BALF sampling for differential cell count and fluorescence-activated cell sorting analysis. RESULTS In this study 26 patients with post-COVID syndrome underwent bronchoscopy with BAL. The HRCT showed ground-glass opacifications (69.2%), organizing pneumonia (7.7%) or both (11.5%). The PFT revealed restrictive lung disease in 38.5% and reduced diffusion capacity in 68%, 19.2% showed a pathological BAL cell pattern predominantly consisting of CD4+ T‑cells. The BALF lymphocyte count was associated with reduced forced vital capacity (p = 0.016) and an elevated alveolar-arterial oxygen gradient (p = 0.04). CONCLUSION A notable percentage of patients with post-COVID syndrome with persistent HRCT changes showed T‑helper lymphocytic inflammation in the lungs. The degree of alveolar lymphocytosis was associated with lung function impairment. This could suggest that a prolonged inflammatory response in the alveolar compartment contributes to the pathogenesis of post-COVID syndrome.
Collapse
Affiliation(s)
- Maximilian Robert Gysan
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Antje Lehmann
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Dominik Bernitzky
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Andreas Zech
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Jonas Brugger
- Institute for Medical Statistics, Medical University of Vienna, Vienna, Austria
| | - Helmut Prosch
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Daniela Gompelmann
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Sauter KA, Webb GM, Bader L, Kreklywich CN, Takahashi DL, Zaro C, McGuire CM, Lewis AD, Colgin LMA, Kirigiti MA, Blomenkamp H, Pessoa C, Humkey M, Hulahan J, Sleeman M, Zweig RC, Thomas S, Thomas A, Gao L, Hirsch AJ, Levy M, Cherry SR, Kahn SE, Slifka MK, Streblow DN, Sacha JB, Kievit P, Roberts CT. Effect of obesity on the acute response to SARS-CoV-2 infection and development of post-acute sequelae of COVID-19 (PASC) in nonhuman primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638792. [PMID: 40027795 PMCID: PMC11870618 DOI: 10.1101/2025.02.18.638792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Long-term adverse consequences of SARS-CoV-2 infection, termed "long COVID" or post-acute sequelae of COVID (PASC), are a major component of overall COVID-19 disease burden. Prior obesity and metabolic disease increase the severity of acute disease, but SARS-CoV-2 infection also contributes to the development of new-onset metabolic disease. Since the COVID pandemic occurred in the context of the global obesity epidemic, an important question is the extent to which pre-existing obesity modifies long-term responses to SARS-CoV-2 infection. We utilized a nonhuman primate model to compare the effects of infection with the SARS-CoV-2 delta variant in lean and obese/insulin-resistant adult male rhesus macaques over a 6-month time course. While some longitudinal responses to SARS-CoV-2 infection, including overall viral dynamics, SARS-CoV-2-specific IgG induction, cytokine profiles, and tissue persistence of viral RNA, did not appreciably differ between lean and obese animals, other responses, including neutralizing Ab dynamics, lung pathology, body weight, degree of insulin sensitivity, adipocytokine profiles, body temperature, and nighttime activity levels were significantly different in lean versus obese animals. Furthermore, several parameters in lean animals were altered following SARS-CoV-2 infection to resemble those in obese animals. Notably, persistent changes in multiple parameters were present in most animals, suggesting that PASC may be more prevalent than estimated from self-reported symptoms in human studies.
Collapse
Affiliation(s)
- Kristin A. Sauter
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
| | | | - Lindsay Bader
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University (OHSU), Beaverton, OR, USA
| | - Diana L. Takahashi
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
| | - Cicely Zaro
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
| | - Casey M. McGuire
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
| | | | | | - Melissa A. Kirigiti
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
| | - Hannah Blomenkamp
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
| | | | | | - Jesse Hulahan
- Department of Pathology and Laboratory Medicine and University of Pennsylvania, Philadelphia, PA, USA
| | - Madeleine Sleeman
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Lina Gao
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Alec. J. Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health and Science University (OHSU), Beaverton, OR, USA
| | - Mayaan Levy
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sara R. Cherry
- Department of Pathology and Laboratory Medicine and University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven E. Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA, USA
| | | | - Daniel N. Streblow
- Division of Pathobiology and Immunology, ONPRC
- Vaccine and Gene Therapy Institute, Oregon Health and Science University (OHSU), Beaverton, OR, USA
| | | | - Paul Kievit
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
| | - Charles T. Roberts
- Division of Metabolic Health and Disease, Oregon National Primate Research Center (ONPRC), Beaverton, OR, USA
- Division of Reproductive and Developmental Sciences, ONPRC
| |
Collapse
|
6
|
Sun DS, Lien TS, Chang HH. Virus-Induced Pathogenic Antibodies: Lessons from Long COVID and Dengue Hemorrhage Fever. Int J Mol Sci 2025; 26:1898. [PMID: 40076527 PMCID: PMC11899886 DOI: 10.3390/ijms26051898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Virus-induced antibodies represent a dual-edged sword in the immune response to viral infections. While antibodies are critical for neutralizing pathogens, some can paradoxically exacerbate disease severity through mechanisms such as antibody-dependent enhancement (ADE), autoantibody, and prolonged inflammation. Long coronavirus disease (COVID) and dengue hemorrhagic fever (DHF) exemplify conditions where pathogenic antibodies play a pivotal role in disease progression. Long COVID is associated with persistent immune dysregulation and autoantibody production, leading to chronic symptoms and tissue damage. In DHF, pre-existing antibodies against dengue virus contribute to ADE, amplifying viral replication, immune activation, and vascular permeability. This review explores the mechanisms underlying these pathogenic antibody responses, highlighting the shared pathways of immune dysregulation and comparing the distinct features of both conditions. By examining these studies, we identify key lessons for therapeutic strategies, vaccine design, and future research aimed at mitigating the severe outcomes of viral infections.
Collapse
Grants
- 104-2320-B-320 -009 -MY3, 107-2311-B-320-002-MY3, 111-2320-B320-006-MY3, 112-2320-B-320-007 National Science and Technology Council, Taiwan
- TCMMP104-06, TCMMP108-04, TCMMP 111-01, TCAS111-02, TCAS-112-02, TCAS113-04, TCRD112-033, TCRD113-041 Tzu-Chi Medical Foundation
Collapse
Affiliation(s)
| | | | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (D.-S.S.); (T.-S.L.)
| |
Collapse
|
7
|
Mahmoud S, Sarkar A, AlMahmoud L, Alladaboina S, Syed LF, Yaghmour M, Elmoh S, AlShebani M, Aly K, Al-Ansari H, Al-Mohamedi M, Yagan L, Zakaria D. Solid Organ Transplants Caused by COVID-19 Infection and the Outcome of Transplantation Post-COVID-19: A Systematic Review. Biomedicines 2025; 13:428. [PMID: 40002841 PMCID: PMC11852956 DOI: 10.3390/biomedicines13020428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic has imposed several medical and economic challenges since its onset in 2019. This is due to its ability to target the respiratory system as well as other organs, resulting in significant impacts and necessitating organ transplants. Our goal is to compile information from the existing literature to investigate how COVID-19 affects outcomes following organ transplantation. A comprehensive literature search was conducted to target studies reporting post-COVID-19 complications. We included 45 studies reporting data related to solid organ transplants, where either the recipient, organ, or donor was affected by SARS-CoV-2. The majority of the included studies concluded that organ transplantation following COVID-19 infection could be performed safely and with similar outcomes to non-COVID-19 patients, regardless of whether the organ, donor, or recipient was affected by COVID-19. No deviation from standard immunosuppression regimens or surgical protocols was necessary either, further re-assuring the feasibility of these transplants as viable treatment options. This applies to organ transplants involving the lungs, kidneys, liver, or heart. However, there was a limited number of studies in some areas, which warrants the need for additional research in order to reach more concrete conclusions pertaining to COVID-19's effect on organ transplants.
Collapse
Affiliation(s)
- Shadi Mahmoud
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Aparajita Sarkar
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Latifa AlMahmoud
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Sushanth Alladaboina
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Leena F. Syed
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Mohammad Yaghmour
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Safaa Elmoh
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Meera AlShebani
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Kareem Aly
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Haya Al-Ansari
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Mohammed Al-Mohamedi
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Lina Yagan
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dalia Zakaria
- Department of Pre-Medical Education, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| |
Collapse
|
8
|
Palladini M, Mazza MG, De Lorenzo R, Spadini S, Aggio V, Bessi M, Calesella F, Bravi B, Rovere-Querini P, Benedetti F. Circulating inflammatory markers predict depressive symptomatology in COVID-19 survivors. Cytokine 2025; 186:156839. [PMID: 39700666 DOI: 10.1016/j.cyto.2024.156839] [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: 11/29/2023] [Revised: 10/30/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Growing evidence suggests the neurobiological mechanism upholding post-COVID-19 depression mainly relates to immune response and subsequent unresolved low-grade inflammation. Herein we exploit a broad panel of cytokines serum levels measured in COVID-19 survivors at one- and three-month since infection to predict post-COVID-19 depression. 87 COVID survivors were screened for depressive symptomatology at one- and three-month after discharge through the Beck Depression Inventory (BDI-13) and the Zung Self-Rating Depression Scale (ZSDS) at San Raffaele Hospital. Blood samples were collected at both timepoints and analyzed through Luminex. We entered one-month 42 inflammatory compounds into two separate penalized logistic regression models to evaluate their reliability in identifying COVID-19 survivors suffering from clinical depression at the two timepoints, applied within a machine learning routine. Delta values of analytes lowering between timepoints were entered in a third model predicting presence long-term depression. 5000 bootstraps were computed to determine significance of predictors. The cross-sectional model reached a balance accuracy (BA) of 76 % and a sensitivity of 70 %. Post-COVID-19 depression was predicted by high levels of CCL17, CCL22. On the other hand, CXCL10, CCL2, CCL3, CCL8, CXCL5, CCL15, CCL23, CXCL13, and GM-CSF showed protective effects. The longitudinal model obtained good performance as well (BA = 74 % and sensitivity = 68 %), revealing CXCL16 and CCL25 as additional drivers of clinical depression. Moreover, dynamic changes of analytes over time accurately predicted long-term depression (BA = 76 % and sensitivity = 75 %). Our findings unveil a putative immune profile upholding post-COVID-19 depression, thus reinforcing the need to deepen molecular mechanisms to appropriately target depression.
Collapse
Affiliation(s)
- Mariagrazia Palladini
- Vita-Salute San Raffaele University, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy.
| | - Mario Gennaro Mazza
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Rebecca De Lorenzo
- School of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Sara Spadini
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Veronica Aggio
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | | | - Federico Calesella
- Vita-Salute San Raffaele University, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Beatrice Bravi
- Vita-Salute San Raffaele University, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Patrizia Rovere-Querini
- School of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy; Unit of Innate Immunity and Tissue Remodeling, Division of Immunology, Transplantation, and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Vita-Salute San Raffaele University, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
| |
Collapse
|
9
|
Aynekulu Mersha DG, Fromme SE, van Boven F, Arteaga-Henríquez G, Wijkhuijs A, van der Ent M, Bergmans R, Baune BT, Drexhage HA, Dalm V. Indications for an antidepressive effect of thymosin alpha-1 in a small open-label proof of concept study in common variable immune deficiency patients with depression. Brain Behav Immun Health 2025; 43:100934. [PMID: 39867848 PMCID: PMC11762651 DOI: 10.1016/j.bbih.2024.100934] [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: 07/20/2024] [Revised: 12/03/2024] [Accepted: 12/21/2024] [Indexed: 01/28/2025] Open
Abstract
Background A considerable proportion (21%) of patients with common variable immunodeficiency (CVID) suffers from depression. These subjects are characterized by reduced naïve T cells and a premature T cell senescence similar to that of patients with major depressive disorder (MDD). It is known that T cells are essential for limbic system development/function. Treatment with thymosin α1 (Tα1) is capable to increase the thymus output of naïve T cells. Objective To treat CVID patients with a comorbid depressive episode with Tα1 to increase naïve T cells and thereby improve mood. Design A small open-label, proof of concept trial. Five depressed CVID patients (Hamilton Depression Rating Scale, HDRS >12) could be treated with Tα1 (8 weeks, 1.6 mg daily subcutaneously, 1st week, thereafter 1.6 mg twice weekly). At the start, at 8 weeks and 8 weeks after the last injection, the HDRS was recorded and blood samples drawn for measuring naïve and memory T cells, Th17 and Treg cells, hsCRP, IL-6 and IL-7. Outcomes were compared to those of a contrast group (42 MDD patients, same severity but treated as usual (TAU)). Results In all 5 depressed CVID patients HDRS decreased during Tα1 treatment (with average 52%, TAU decreased scores with 36% in MDD patients). All 5 CVID patients showed an increase in naïve/memory CD4+ and CD8+ T cell ratios, and in 4 of the 5 patients with detectable IL-6 levels reductions were recorded. TAU did not show such immune improvements. In the 8-week wash-out, depression recurred in the 2 most severe patients, while continued to improve in the others. Immune effects were not sustained in the wash-out. Conclusion This preliminary small study suggests thymus hormone treatment to have antidepressive and related immune correcting effects. Data urge for larger placebo-controlled trials.
Collapse
Affiliation(s)
| | - Sarah E. Fromme
- Dept of Psychiatry, University of Muenster, Muenster, Germany
| | - Frank van Boven
- Department of Internal Medicine, Section of Allergology & Clinical Immunology, Erasmus MC, University Medical Center Rotterdam, P.O. Box 2040, 3000, CA, Rotterdam, the Netherlands
| | - Gara Arteaga-Henríquez
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Catalonia, Spain
- Group of Psychiatry, Mental Health, and Addictions, Vall d'Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain
- Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain
| | | | | | - Raf Bergmans
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
| | - Bernard T. Baune
- Dept of Psychiatry, University of Muenster, Muenster, Germany
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
| | - Hemmo A. Drexhage
- Dept of Immunology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Virgil Dalm
- Dept of Immunology, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
10
|
Chaves ECR, Quaresma JAS, Rodrigues MHC, de Menezes DC, de Lima IC, de Sousa JR, Galúcio VCA, Queiroz MAF, Brasil-Costa I, Barros MC, Ribeiro-Dos-Santos Â, Vallinoto ACR, Falcão LFM, de Lima PDL. Altered leukocyte pattern and inflammatory markers in unvaccinated long covid patients: a cross-sectional study. Sci Rep 2024; 14:28617. [PMID: 39562810 PMCID: PMC11577114 DOI: 10.1038/s41598-024-75920-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/09/2024] [Indexed: 11/21/2024] Open
Abstract
Long Covid results from the damage caused by SARS-CoV-2, involving the release of cytokines and the continuous activation of immune cells. This cross-sectional study investigates leukocyte and cytokine profiles in Long Covid patients in the Amazon, a region where such studies are limited. Blood samples were analysed for differential leukocyte counts and cytokine levels. We suggest elevated lymphocyte counts in hospitalised patients and those with severe COVID-19. Higher eosinophil counts were observed in patients with up to three months of Long Covid, and increased monocyte counts in those with up to six months. IL-2 levels were higher in patients with fewer symptoms and Long Covid duration of more than three months, whereas IL-10 may remain elevated for up to 12 months. We suggest positive correlations between neutrophils, monocytes, eosinophils, and lymphocytes with different cytokines (IFN-γ, IL-6, IL-4, IL-17a, IL-2). Women were associated with lower hospitalisation rates and longer durations of Long Covid; increased lymphocyte counts were linked to hospitalisation due to COVID-19, while higher monocyte counts were associated with Long Covid durations of up to six months. We suggest that Long Covid patients may exhibit alterations in inflammatory markers, indicating a persistently pro-inflammatory microenvironment that tends to diminish after 12 months of Long Covid.
Collapse
Affiliation(s)
- Elem Cristina Rodrigues Chaves
- Department of Center for Biological Health Sciences (CCBS), Graduate Program in Parasitic Biology in Amazonia (PPGBPA), State University of Pará (UEPA), Marco, Belém, Pará, 66087-670, Brazil
| | - Juarez Antônio Simões Quaresma
- Department of Center for Biological Health Sciences (CCBS), Graduate Program in Parasitic Biology in Amazonia (PPGBPA), State University of Pará (UEPA), Marco, Belém, Pará, 66087-670, Brazil
| | | | - Daniel Carvalho de Menezes
- Department of Center for Biological Health Sciences (CCBS), Graduate Program in Parasitic Biology in Amazonia (PPGBPA), State University of Pará (UEPA), Marco, Belém, Pará, 66087-670, Brazil
| | - Igor Costa de Lima
- Department of Center for Biological Health Sciences (CCBS), Graduate Program in Parasitic Biology in Amazonia (PPGBPA), State University of Pará (UEPA), Marco, Belém, Pará, 66087-670, Brazil
| | - Jorge Rodrigues de Sousa
- Department of Morphology and Physiological Sciences (DMCF), State University of Pará (UEPA), Belém, Pará, 66087- 670, Brazil
| | - Vanessa Costa Alves Galúcio
- Department of Center for Biological Health Sciences (CCBS), Graduate Program in Parasitic Biology in Amazonia (PPGBPA), State University of Pará (UEPA), Marco, Belém, Pará, 66087-670, Brazil
| | - Maria Alice Freitas Queiroz
- Laboratory of Virology (LABVIR), Institute of Biological Sciences (ICB), Federal University of Pará (UFPA), Belém, Pará, 66075-110, Brazil
| | - Igor Brasil-Costa
- Immunology Laboratory, Virology Section, Evandro Chagas Institute (IEC), Health and Environmental Surveillance Secretariat (SVSA), Brazilian Ministry of Health, Ananindeua, Brazil
| | - Maria Clara Barros
- Laboratory of Human and Medical Genetics (LGHM), Federal University of Pará (UFPA), Belém, Pará, 66075-110, Brazil
| | - Ândrea Ribeiro-Dos-Santos
- Laboratory of Human and Medical Genetics (LGHM), Federal University of Pará (UFPA), Belém, Pará, 66075-110, Brazil
| | - Antonio Carlos Rosário Vallinoto
- Laboratory of Virology (LABVIR), Institute of Biological Sciences (ICB), Federal University of Pará (UFPA), Belém, Pará, 66075-110, Brazil
| | - Luiz Fábio Magno Falcão
- Department of Center for Biological Health Sciences (CCBS), Graduate Program in Parasitic Biology in Amazonia (PPGBPA), State University of Pará (UEPA), Marco, Belém, Pará, 66087-670, Brazil
- School of Medicine, São Paulo University (USP), São Paulo, 01246903, Brazil
| | - Patrícia Danielle Lima de Lima
- Department of Center for Biological Health Sciences (CCBS), Graduate Program in Parasitic Biology in Amazonia (PPGBPA), State University of Pará (UEPA), Marco, Belém, Pará, 66087-670, Brazil.
| |
Collapse
|
11
|
Elliott MR, O'Connor AE, Marshall GD. Inflammatory pathways in patients with post-acute sequelae of COVID-19: The role of the clinical immunologist. Ann Allergy Asthma Immunol 2024; 133:507-515. [PMID: 39179099 PMCID: PMC11575468 DOI: 10.1016/j.anai.2024.08.021] [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: 05/30/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
As the SARS-CoV-2 pandemic progressed, some survivors noted prolonged symptoms after acute infection, termed post-acute sequelae of COVID-19 (PASC) or "long COVID." PASC is a significant clinical and public health concern that adversely affects patients' quality of life, income, and health care expenses. Moreover, PASC symptoms are highly heterogeneous, the most common being fatigue and cognitive impairment, and they likely reflect a spectrum of clinical phenotypes. The proposed role of persistent inflammation is one of leading pathophysiological theories. This review article addresses these proposed mechanisms of persistent and aberrant inflammation, their clinical evaluation, and theoretical approaches to management. A review of public databases was used to collect literature for the review. The literature supports a prominent role of persistent and aberrant inflammation as a major contributor to the symptoms of PASC. Proposed mechanisms for persistent inflammation include reactivation of latent viruses, viral persistence, loss of immunoregulatory pathways, autoimmune mechanisms, and/or mast cell dysregulation. Persistent inflammation may result in constitutional symptoms such as fatigue, brain fog, body aches, and/or organ-specific dysfunction, such as gastrointestinal dysregulation and myocardial inflammation. There are no approved or even proven therapies for PASC at this time, but some studies have identified therapeutic options that may either reduce the risk for progression to PASC or decrease symptom burden. Laboratory evaluation and therapeutic options are limited and require further investigation to establish their clinical value. A more refined definition of PASC is needed to address the wide variety of clinical presentations, pathophysiology, and therapeutic options.
Collapse
Affiliation(s)
- Matthew R Elliott
- The University of Mississippi Medical Center, Department of Internal Medicine, Division of Clinical Immunology, Jackson, Mississippi.
| | - Anna E O'Connor
- The University of Mississippi Medical Center, Department of Internal Medicine, Division of Clinical Immunology, Jackson, Mississippi
| | - Gailen D Marshall
- The University of Mississippi Medical Center, Department of Internal Medicine, Division of Clinical Immunology, Jackson, Mississippi
| |
Collapse
|
12
|
Poethko-Müller C, Schaffrath Rosario A, Sarganas G, Ordonez Cruickshank A, Scheidt-Nave C, Schlack R. [Fatigue in the general population: results of the "German Health Update 2023" study]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2024; 67:1208-1221. [PMID: 39327264 PMCID: PMC11549105 DOI: 10.1007/s00103-024-03950-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Fatigue is an unspecific symptom complex characterized by tiredness, lack of energy, and lack of concentration and is of considerable public health relevance, due to its links with incapacity for work, risk of accidents, and increased need for healthcare. METHODS The analyses are based on data from 9766 adults of the telephone survey "Gesundheit in Deutschland aktuell (GEDA)" 2023. Fatigue was recorded using the Fatigue Assessment Scale (FAS), a validated instrument with 10 questions for self-assessment of fatigue. The scale was dichotomized into yes (at least mild to moderate fatigue) versus no (no fatigue). Population-weighted prevalences of fatigue and associated sociodemographic and health-related factors were calculated in descriptive analyses and multivariable Poisson regression. RESULTS The overall prevalence of fatigue in adults in Germany is 29.7% (95% CI 28.1-31.2), is highest in 18- to 29-year-olds (39.6% (95% CI 35.0-44.4)), and decreases in the age groups up to 65-79 years (20.6% (95% CI 18.2-23.3)). It is higher again in the very old age group (33.2% (95% CI 28.9-37.7)). Women have a higher risk of fatigue than men (aRR 1.19 (95% CI 1.08-1.32)). Fatigue is significantly associated with age, lower education, chronic illness, depression, and long COVID, regardless of covariates. DISCUSSION GEDA 2023 is one of the few population-based studies to have collected data on fatigue. The results allow estimates to be made for Germany on the frequency of fatigue and the significance of physical, psychological, and social influencing factors. They can be used as a reference or as a basis for trends over time as part of continuous health monitoring in Germany.
Collapse
Affiliation(s)
- Christina Poethko-Müller
- Abt. Epidemiologie und Gesundheitsmonitoring, FG Körperliche Gesundheit, Robert Koch-Institut, General-Pape-Str. 62-66, 12101, Berlin, Deutschland.
| | - Angelika Schaffrath Rosario
- Abt. Epidemiologie und Gesundheitsmonitoring, FG Gesundheitsberichterstattung, Robert Koch-Institut, Berlin, Germany
| | - Giselle Sarganas
- Abt. Epidemiologie und Gesundheitsmonitoring, FG Körperliche Gesundheit, Robert Koch-Institut, General-Pape-Str. 62-66, 12101, Berlin, Deutschland
| | - Ana Ordonez Cruickshank
- Abt. Epidemiologie und Gesundheitsmonitoring, FG Körperliche Gesundheit, Robert Koch-Institut, General-Pape-Str. 62-66, 12101, Berlin, Deutschland
| | - Christa Scheidt-Nave
- Abt. Epidemiologie und Gesundheitsmonitoring, FG Körperliche Gesundheit, Robert Koch-Institut, General-Pape-Str. 62-66, 12101, Berlin, Deutschland
| | - Robert Schlack
- Abt. Epidemiologie und Gesundheitsmonitoring, FG Psychische Gesundheit, Robert Koch-Institut, Berlin, Germany
| |
Collapse
|
13
|
Almulla AF, Thipakorn Y, Zhou B, Vojdani A, Maes M. Immune activation and immune-associated neurotoxicity in Long-COVID: A systematic review and meta-analysis of 103 studies comprising 58 cytokines/chemokines/growth factors. Brain Behav Immun 2024; 122:75-94. [PMID: 39127088 DOI: 10.1016/j.bbi.2024.07.036] [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: 04/10/2024] [Revised: 07/18/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Multiple studies have shown that Long COVID (LC) disease is associated with heightened immune activation, as evidenced by elevated levels of inflammatory mediators. However, there is no comprehensive meta-analysis focusing on activation of the immune inflammatory response system (IRS) and the compensatory immunoregulatory system (CIRS) along with other immune phenotypes in LC patients. OBJECTIVES This meta-analysis is designed to explore the IRS and CIRS profiles in LC patients, the individual cytokines, chemokines, growth factors, along with C-reactive protein (CRP) and immune-associated neurotoxicity. METHODS To gather relevant studies for our research, we conducted a thorough search using databases such as PubMed, Google Scholar, and SciFinder, covering all available literature up to July 5th, 2024. RESULTS The current meta-analysis encompassed 103 studies that examined multiple immune profiles, C-reactive protein, and 58 cytokines/chemokines/growth factors in 5502 LC patients versus 5962 normal controls (NC). LC patients showed significant increases in IRS/CIRS ratio (standardized mean difference (SMD: 0.156, confidence interval (CI): 0.062;0.250), IRS (SMD: 0.338, CI: 0.236;0.440), M1 macrophage (SMD: 0.371, CI: 0.263;0.480), T helper (Th)1 (SMD: 0.316, CI: 0.185;0.446), Th17 (SMD: 0.439, CI: 0.302;0.577) and immune-associated neurotoxicity (SMD: 0.384, CI: 0.271;0.497). In addition, CRP and 21 different cytokines displayed significantly elevated levels in LC patients compared to NC. CONCLUSION LC disease is characterized by IRS activation and increased immune-associated neurotoxicity.
Collapse
Affiliation(s)
- Abbas F Almulla
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Yanin Thipakorn
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Bo Zhou
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China
| | - Aristo Vojdani
- Immunosciences Lab, Inc., Los Angeles, CA 90035, USA; Cyrex Laboratories, LLC, Phoenix, AZ 85034, USA
| | - Michael Maes
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Fitness and Biopsychological Technology Research Unit, Faculty of Medicine. Chulalongkorn University, Bangkok 10330, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria; Strategic Research and Innovation Program for the Development of MU - PLOVDIV-(SRIPD-MUP), European Union - NextGenerationEU; Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, South Korea.
| |
Collapse
|
14
|
Gao J, Zhang C, Wheelock ÅM, Xin S, Cai H, Xu L, Wang XJ. Immunomics in one health: understanding the human, animal, and environmental aspects of COVID-19. Front Immunol 2024; 15:1450380. [PMID: 39295871 PMCID: PMC11408184 DOI: 10.3389/fimmu.2024.1450380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 09/21/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic underscores the critical need to integrate immunomics within the One Health framework to effectively address zoonotic diseases across humans, animals, and environments. Employing advanced high-throughput technologies, this interdisciplinary approach reveals the complex immunological interactions among these systems, enhancing our understanding of immune responses and yielding vital insights into the mechanisms that influence viral spread and host susceptibility. Significant advancements in immunomics have accelerated vaccine development, improved viral mutation tracking, and broadened our comprehension of immune pathways in zoonotic transmissions. This review highlights the role of animals, not merely as carriers or reservoirs, but as essential elements of ecological networks that profoundly influence viral epidemiology. Furthermore, we explore how environmental factors shape immune response patterns across species, influencing viral persistence and spillover risks. Moreover, case studies demonstrating the integration of immunogenomic data within the One Health framework for COVID-19 are discussed, outlining its implications for future research. However, linking humans, animals, and the environment through immunogenomics remains challenging, including the complex management of vast amounts of data and issues of scalability. Despite challenges, integrating immunomics data within the One Health framework significantly enhances our strategies and responses to zoonotic diseases and pandemic threats, marking a crucial direction for future public health breakthroughs.
Collapse
Affiliation(s)
- Jing Gao
- Department of Respiratory Medicine, Gansu Provincial Hospital, Lanzhou, China
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Pulmonary Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Chutian Zhang
- College of Natural Resources and Environment, Northwest Agriculture and Forestry University, Yangling, China
| | - Åsa M Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Siming Xin
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Hui Cai
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
| | - Lei Xu
- Vanke School of Public Health, Tsinghua University, Beijing, China
- Institute for Healthy China, Tsinghua University, Beijing, China
| | - Xiao-Jun Wang
- Department of Respiratory Medicine, Gansu Provincial Hospital, Lanzhou, China
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
15
|
Thomas C, Faghy MA, Chidley C, Phillips BE, Bewick T, Ashton RE. Blood Biomarkers of Long COVID: A Systematic Review. Mol Diagn Ther 2024; 28:537-574. [PMID: 39103645 DOI: 10.1007/s40291-024-00731-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Long coronavirus disease (COVID; LC) affects millions of people worldwide. The exact mechanisms which result in a broad, undulating and detrimental symptom profile remain unknown. Blood biomarkers associated with LC have been described; however, consensus on these remains elusive, in part due to a lack of continuity between studies on a universally accepted definition of LC. This systematic review aimed to consolidate current knowledge of blood biomarkers associated with the prevalence of LC on the basis of the World Health Organisation (WHO) clinical definition of this condition. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Observational, cross-sectional, and randomised control studies published in the English language that studied blood biomarkers associated with the WHO definition of LC. All studies included participants who were ≥ 18 years old and group sizes ≥ 10 participants, and were compared against a control group without any known co-morbidities. METHODS A systematic literature search was conducted according to Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines and prospectively registered on Prospero (ID: CRD42022373121). The Cochrane, Embase, PubMed and Web of Science databases were searched from inception to January 2024. Search results were gathered using Rayyan software and data extracted using Microsoft Excel. The reporting recommendations for tumour markers prognostic studies (REMARK) questionnaire was used to assess the quality of the included studies. RESULTS A total of 45 observational and one interventional study comprising 4415 participants were included in this review which identified 525 blood biomarkers thought to be associated with LC. Three blood biomarker subtypes were associated with the development of LC: (1) immunological and inflammatory dysfunction, (2) endothelial/vascular dysfunction and (3) metabolic and clotting abnormalities. DISCUSSION AND CONCLUSIONS Our data are consistent with previous findings; however, no single biomarker was sufficiently associated with LC prevalence and instead a profile of biomarkers across various physiological systems may be more clinically useful. In all, 196 studies were excluded due to a lack of an adequately healthy comparator group and/or failure to meet the WHO LC definition. This demonstrates a need for further research incorporating a universal LC definition across all disease severity groups and symptom profiles, and longitudinal data reflecting the relapsing and remitting nature of this condition. Further investigation into blood biomarkers of LC, including clear reporting of healthy comparator groups and the investigation of acute and chronic biomarker changes, within the context of medical practice, may support the development of curative/restorative approaches.
Collapse
Affiliation(s)
- Callum Thomas
- Biomedical and Clinical Science Research Theme, School of Human Sciences, University of Derby, Derby, UK.
- Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA.
| | - Mark A Faghy
- Biomedical and Clinical Science Research Theme, School of Human Sciences, University of Derby, Derby, UK
- Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA
- Department of Physical Therapy, College of Applied Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Corinna Chidley
- Biomedical and Clinical Science Research Theme, School of Human Sciences, University of Derby, Derby, UK
| | - Bethan E Phillips
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Thomas Bewick
- Department of Respiratory Medicine, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| | - Ruth E Ashton
- Biomedical and Clinical Science Research Theme, School of Human Sciences, University of Derby, Derby, UK
- Healthy Living for Pandemic Event Protection (HL-PIVOT) Network, Chicago, IL, USA
- Research Centre for Physical Activity, Sport and Exercise Sciences, Coventry University, Coventry, UK
| |
Collapse
|
16
|
Liu C, Ge P, Zhang B, Chan L, Pang Y, Tao C, Li J, He Q, Liu W, Mou S, Zheng Z, Zhao Z, Sun W, Zhang Q, Wang R, Zhang Y, Wang W, Zhang D, Zhao J. Mass cytometry revealed the circulating immune cell landscape across different Suzuki stages of Moyamoya disease. Immunol Res 2024; 72:654-664. [PMID: 38376705 PMCID: PMC11347468 DOI: 10.1007/s12026-024-09464-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/04/2024] [Indexed: 02/21/2024]
Abstract
Moyamoya disease (MMD) is a cerebrovascular disorder marked by progressive arterial narrowing, categorized into six stages known as Suzuki stages based on angiographic features. Growing evidence indicates a pivotal role of systemic immune and inflammatory responses in the initiation and advancement of MMD. This study employs high-dimensional mass cytometry to reveal the immunophenotypic characteristics of peripheral blood immune cells (PBMCs) at various Suzuki stages, offering insights into the progression of MMD. PBMC samples from eight patients with early-stage MMD (Suzuki stages II and III) and eight patients with later-stage MMD (Suzuki stages IV, V, and VI) were analyzed using high-dimensional mass cytometry to evaluate the frequency and phenotype of immune cell subtypes. We identified 15 cell clusters and found that the immunological features of early-stage MMD and later-stage MMD are composed of cluster variations. In this study, we confirmed that, compared to later-stage MMD, the early-stage MMD group exhibits an increase in non-classical monocytes. As the Suzuki stage level increases, the proportions of plasmacytoid DCs and monocyte-derived DCs decrease. Furthermore, T cells, monocytes, DCs, and PMN-MDSCs in the early-stage MMD group show activation of the canonical NF-κB signaling pathway. We summarized and compared the similarities and differences between early-stage MMD patients and later-stage MMD patients. There is a potential role of circulating immune dysfunction and inflammatory responses in the onset and development of MMD.
Collapse
Affiliation(s)
- Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Bojian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Liujia Chan
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Yuheng Pang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China
| | - Chuming Tao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Wei Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Siqi Mou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Zhiyao Zheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Zhikang Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Wei Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China.
| | - Dong Zhang
- Department of Neurosurgery, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China.
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China.
| |
Collapse
|
17
|
Campisi M, Cannella L, Bordin A, Moretto A, Scapellato ML, Mason P, Liviero F, Pavanello S, on behalf of Occupational Medicine Working Group. Revealing the Hidden Impacts: Insights into Biological Aging and Long-Term Effects in Pauci- and Asymptomatic COVID-19 Healthcare Workers. Int J Mol Sci 2024; 25:8056. [PMID: 39125624 PMCID: PMC11311509 DOI: 10.3390/ijms25158056] [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: 07/02/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
This study explores the role of inflammation and oxidative stress, hallmarks of COVID-19, in accelerating cellular biological aging. We investigated early molecular markers-DNA methylation age (DNAmAge) and telomere length (TL)-in blood leukocytes, nasal cells (NCs), and induced sputum (IS) one year post-infection in pauci- and asymptomatic healthcare workers (HCWs) infected during the first pandemic wave (February-May 2020), compared to COPD patients, model for "aged lung". Data from questionnaires, Work Ability Index (WAI), blood analyses, autonomic cardiac balance assessments, heart rate variability (HRV), and pulmonary function tests were collected. Elevated leukocyte DNAmAge significantly correlated with advancing age, male sex, daytime work, and an aged phenotype characterized by chronic diseases, elevated LDL and glycemia levels, medications affecting HRV, and declines in lung function, WAI, lymphocyte count, hemoglobin levels, and HRV (p < 0.05). Increasing age, LDL levels, job positions involving intensive patient contact, and higher leukocyte counts collectively contributed to shortened leukocyte TL (p < 0.05). Notably, HCWs exhibited accelerated biological aging in IS cells compared to both blood leukocytes (p ≤ 0.05) and NCs (p < 0.001) and were biologically older than COPD patients (p < 0.05). These findings suggest the need to monitor aging in pauci- and asymptomatic COVID-19 survivors, who represent the majority of the general population.
Collapse
Affiliation(s)
- Manuela Campisi
- Department of Cardiac-, -Thoracic-, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy; (M.C.); (L.C.); (A.B.); (A.M.); (M.L.S.); (P.M.); (F.L.)
| | - Luana Cannella
- Department of Cardiac-, -Thoracic-, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy; (M.C.); (L.C.); (A.B.); (A.M.); (M.L.S.); (P.M.); (F.L.)
| | - Anna Bordin
- Department of Cardiac-, -Thoracic-, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy; (M.C.); (L.C.); (A.B.); (A.M.); (M.L.S.); (P.M.); (F.L.)
- Occupational Medicine, University Hospital of Padua, 35128 Padua, Italy
| | - Angelo Moretto
- Department of Cardiac-, -Thoracic-, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy; (M.C.); (L.C.); (A.B.); (A.M.); (M.L.S.); (P.M.); (F.L.)
- Occupational Medicine, University Hospital of Padua, 35128 Padua, Italy
| | - Maria Luisa Scapellato
- Department of Cardiac-, -Thoracic-, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy; (M.C.); (L.C.); (A.B.); (A.M.); (M.L.S.); (P.M.); (F.L.)
- Occupational Medicine, University Hospital of Padua, 35128 Padua, Italy
| | - Paola Mason
- Department of Cardiac-, -Thoracic-, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy; (M.C.); (L.C.); (A.B.); (A.M.); (M.L.S.); (P.M.); (F.L.)
- Occupational Medicine, University Hospital of Padua, 35128 Padua, Italy
| | - Filippo Liviero
- Department of Cardiac-, -Thoracic-, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy; (M.C.); (L.C.); (A.B.); (A.M.); (M.L.S.); (P.M.); (F.L.)
- Occupational Medicine, University Hospital of Padua, 35128 Padua, Italy
| | - Sofia Pavanello
- Department of Cardiac-, -Thoracic-, Vascular Sciences and Public Health, University of Padua, 35128 Padua, Italy; (M.C.); (L.C.); (A.B.); (A.M.); (M.L.S.); (P.M.); (F.L.)
- Occupational Medicine, University Hospital of Padua, 35128 Padua, Italy
| | | |
Collapse
|
18
|
Bhuvaneshwar K, Madhavan S, Gusev Y. Integrative genomic analysis of the lung tissue microenvironment in SARS-CoV-2 and NL63 patients. Heliyon 2024; 10:e32772. [PMID: 39183848 PMCID: PMC11341340 DOI: 10.1016/j.heliyon.2024.e32772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/28/2024] [Accepted: 06/09/2024] [Indexed: 08/27/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-CoV-2 virus has affected over 700 million people, and caused over 7 million deaths throughout the world as of April 2024, and continues to affect people through seasonal waves. While over 675 million people have recovered from this disease globally, the lingering effects of the disease are still under study. Long term effects of SARS-CoV-2 infection, known as 'long COVID,' include a wide range of symptoms including fatigue, chest pain, cellular damage, along with a strong innate immune response characterized by inflammatory cytokine production. Three years after the pandemic, data about long covid studies are finally emerging. More clinical studies and clinical trials are needed to understand and determine the factors that predispose individuals to these long-term side effects. In this methodology paper, our goal was to apply data driven approaches in order to explore the multidimensional landscape of infected lung tissue microenvironment to better understand complex interactions between viral infection, immune response and the lung microbiome of patients with (a) SARS-CoV-2 virus and (b) NL63 coronavirus. The samples were analyzed with several machine learning tools allowing simultaneous detection and quantification of viral RNA amount at genome and gene level; human gene expression and fractions of major types of immune cells, as well as metagenomic analysis of bacterial and viral abundance. To contrast and compare specific viral response to SARS-COV-2, we analyzed deep sequencing data from additional cohort of patients infected with NL63 strain of corona virus. Our correlation analysis of three types of RNA-seq based measurements in patients i.e. fraction of viral RNA (at genome and gene level), Human RNA (transcripts and gene level) and bacterial RNA (metagenomic analysis), showed significant correlation between viral load as well as level of specific viral gene expression with the fractions of immune cells present in lung lavage as well as with abundance of major fractions of lung microbiome in COVID-19 patients. Our methodology-based proof-of-concept study has provided novel insights into complex regulatory signaling interactions and correlative patterns between the viral infection, inhibition of innate and adaptive immune response as well as microbiome landscape of the lung tissue. These initial findings could provide better understanding of the diverse dynamics of immune response and the side effects of the SARS-CoV-2 infection and demonstrates the possibilities of the various types of analyses that could be performed from this type of data.
Collapse
Affiliation(s)
- Krithika Bhuvaneshwar
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| | - Subha Madhavan
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| | - Yuriy Gusev
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| |
Collapse
|
19
|
Gusev E, Sarapultsev A. Exploring the Pathophysiology of Long COVID: The Central Role of Low-Grade Inflammation and Multisystem Involvement. Int J Mol Sci 2024; 25:6389. [PMID: 38928096 PMCID: PMC11204317 DOI: 10.3390/ijms25126389] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Long COVID (LC), also referred to as Post COVID-19 Condition, Post-Acute Sequelae of SARS-CoV-2 Infection (PASC), and other terms, represents a complex multisystem disease persisting after the acute phase of COVID-19. Characterized by a myriad of symptoms across different organ systems, LC presents significant diagnostic and management challenges. Central to the disorder is the role of low-grade inflammation, a non-classical inflammatory response that contributes to the chronicity and diversity of symptoms observed. This review explores the pathophysiological underpinnings of LC, emphasizing the importance of low-grade inflammation as a core component. By delineating the pathogenetic relationships and clinical manifestations of LC, this article highlights the necessity for an integrated approach that employs both personalized medicine and standardized protocols aimed at mitigating long-term consequences. The insights gained not only enhance our understanding of LC but also inform the development of therapeutic strategies that could be applicable to other chronic conditions with similar pathophysiological features.
Collapse
Affiliation(s)
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia;
| |
Collapse
|
20
|
Li W, Huang X, Wei Y, Yin T, Diao L. Connecting the dots: the role of fatigue in female infertility. Reprod Biol Endocrinol 2024; 22:66. [PMID: 38849828 PMCID: PMC11157719 DOI: 10.1186/s12958-024-01235-5] [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: 01/30/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
Fatigue, an increasingly acknowledged symptom in various chronic diseases, has garnered heightened attention, during the medical era of bio-psycho-social model. Its persistence not only significantly compromises an individual's quality of life but also correlates with chronic organ damage. Surprisingly, the intricate relationship between fatigue and female reproductive health, specifically infertility, remains largely unexplored. Our exploration into the existing body of evidence establishes a compelling link between fatigue with uterine and ovarian diseases, as well as conditions associated with infertility, such as rheumatism. This observation suggests a potentially pivotal role of fatigue in influencing overall female fertility. Furthermore, we propose a hypothetical mechanism elucidating the impact of fatigue on infertility from multiple perspectives, postulating that neuroendocrine, neurotransmitter, inflammatory immune, and mitochondrial dysfunction resulting from fatigue and its co-factors may further contribute to endocrine disorders, menstrual irregularities, and sexual dysfunction, ultimately leading to infertility. In addition to providing this comprehensive theoretical framework, we summarize anti-fatigue strategies and accentuate current knowledge gaps. By doing so, our aim is to offer novel insights, stimulate further research, and advance our understanding of the crucial interplay between fatigue and female reproductive health.
Collapse
Grants
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
Collapse
Affiliation(s)
- Wenzhu Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Xiaoyan Huang
- Department of Rheumatology, The University of Hong Kong- Shenzhen Hospital, Shenzhen, 518053, China
| | - Yiqiu Wei
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China.
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518045, China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri- implantation, Shenzhen, 518045, China.
| |
Collapse
|
21
|
Berentschot JC, Martine Bek L, Heijenbrok-Kal MH, van den Berg-Emons RJG, Ribbers GM, Aerts JGJV, Hellemons ME. Acute COVID-19 treatment is not associated with health problems 2 years after hospitalization. Int J Infect Dis 2024; 142:106966. [PMID: 38367953 DOI: 10.1016/j.ijid.2024.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024] Open
Abstract
OBJECTIVES Various mechanisms, such as immune dysregulation, viral reservoir, and auto-immunity, are hypothesized to underlie the pathogenesis of long-term health problems after hospitalization for COVID-19. We aimed to assess the effect of in-hospital COVID-19 treatments on prominent long-term health problems. METHODS In this prospective multicenter cohort study, we enrolled patients (age ≥18 years) who had been hospitalized for COVID-19 in the Netherlands between July 2020 and October 2021. We retrospectively collected data on in-hospital COVID-19 treatments, including steroid, anti-inflammatory, and antiviral treatments. Patients completed questionnaires on self-reported recovery, dyspnea, fatigue, cognitive failures, and health-related quality of life and performed the 6-minute walk test at the 2-year follow-up visit. RESULTS Five hundred two patients with COVID-19 were included, all were discharged from the hospital between March 2020 and June 2021. The median age at admission was 60.0 (IQR 53.0-68.0) years and 350 (69.7%) patients were male. At hospital admission, 5/405 (1.2%) of the patients had been vaccinated against SARS-CoV-2. Among all 502 patients, the majority (248 [49.4%]) received steroids only, 57 (11.4%) anti-inflammatory treatment, 78 (15.5%) antiviral treatment, and 119 (23.7%) none during hospitalization. Long-term health problems were common in all groups. We found that in-hospital treatments were not significantly associated with health problems at 2 years after hospital discharge, nor after adjusting for confounders. CONCLUSION Many patients with COVID-19 suffer from long-term health problems 2 years after hospital discharge. Acute treatment for COVID-19 is not associated with long-term health problems.
Collapse
Affiliation(s)
- Julia C Berentschot
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - L Martine Bek
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Majanka H Heijenbrok-Kal
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Rijndam Rehabilitation, Rotterdam, The Netherlands
| | - Rita J G van den Berg-Emons
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gerard M Ribbers
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Rijndam Rehabilitation, Rotterdam, The Netherlands
| | - Joachim G J V Aerts
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Merel E Hellemons
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Giunta S, Giordani C, De Luca M, Olivieri F. Long-COVID-19 autonomic dysfunction: An integrated view in the framework of inflammaging. Mech Ageing Dev 2024; 218:111915. [PMID: 38354789 DOI: 10.1016/j.mad.2024.111915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
The recently identified syndrome known as Long COVID (LC) is characterized by a constellation of debilitating conditions that impair both physical and cognitive functions, thus reducing the quality of life and increasing the risk of developing the most common age-related diseases. These conditions are linked to the presence of symptoms of autonomic dysfunction, in association with low cortisol levels, suggestive of reduced hypothalamic-pituitary-adrenal (HPA) axis activity, and with increased pro-inflammatory condition. Alterations of dopamine and serotonin neurotransmitter levels were also recently observed in LC. Interestingly, at least some of the proposed mechanisms of LC development overlap with mechanisms of Autonomic Nervous System (ANS) imbalance, previously detailed in the framework of the aging process. ANS imbalance is characterized by a proinflammatory sympathetic overdrive, and a concomitant decreased anti-inflammatory vagal parasympathetic activity, associated with reduced anti-inflammatory effects of the HPA axis and cholinergic anti-inflammatory pathway (CAP). These neuro-immune-endocrine system imbalanced activities fuel the vicious circle of chronic inflammation, i.e. inflammaging. Here, we refine our original hypothesis that ANS dysfunction fuels inflammaging and propose that biomarkers of ANS imbalance could also be considered biomarkers of inflammaging, recognized as the main risk factor for developing age-related diseases and the sequelae of viral infections, i.e. LC.
Collapse
Affiliation(s)
- Sergio Giunta
- Casa di Cura Prof. Nobili (Gruppo Garofalo (GHC) Castiglione dei Pepoli -Bologna), Italy
| | - Chiara Giordani
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy.
| | - Maria De Luca
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Fabiola Olivieri
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy; Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
23
|
Sommen SL, Zhao Z, Segtnan S, Stiansen-Sonerud T, Selvakumar J, Beier Havdal L, Gjerstad J, Wyller VBB, Lund Berven L. Bulk RNA sequencing for analysis of post COVID-19 condition in adolescents and young adults. J Transl Med 2024; 22:312. [PMID: 38532465 PMCID: PMC10964710 DOI: 10.1186/s12967-024-05117-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Post COVID-19 condition (PCC) is a complication of SARS-COV-2 infection and can lead to long-term disability. METHODS The present study was designed to analyse the gene expression patterns of PCC through bulk RNA sequencing of whole blood and to explore the potential molecular mechanisms of PCC. Whole blood was collected from 80 participants enrolled in a prospective cohort study following SARS-CoV-2 infected and non-infected individuals for 6 months after recruitment and was used for bulk RNA sequencing. Identification of differentially expressed genes (DEG), pathway enrichment and immune cell deconvolution was performed to explore potential biological pathways involved in PCC. RESULTS We have found 13 differentially expressed genes associated with PCC. Enriched pathways were related to interferon-signalling and anti-viral immune processes. CONCLUSION The PCC transcriptome is characterized by a modest overexpression of interferon-stimulated genes, pointing to a subtle ongoing inflammatory response.
Collapse
Affiliation(s)
- Silke Lauren Sommen
- Department of Pediatrics, Akershus University Hospital, Lørenskog, Norway
- University of Oslo, Oslo, Norway
| | - Zhi Zhao
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Tonje Stiansen-Sonerud
- Department of Pediatrics, Akershus University Hospital, Lørenskog, Norway
- Department of Clinical Molecular Biology (EpiGen), University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Joel Selvakumar
- Department of Pediatrics, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lise Beier Havdal
- Department of Pediatrics, Akershus University Hospital, Lørenskog, Norway
| | - Johannes Gjerstad
- Department of Behavioural Sciences, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| | - Vegard Bruun Bratholm Wyller
- Department of Pediatrics, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lise Lund Berven
- Department of Pediatrics, Akershus University Hospital, Lørenskog, Norway.
| |
Collapse
|
24
|
Quinn J. Long COVID autonomic syndrome: Improved understanding through translational research. Eur J Intern Med 2024; 120:34-35. [PMID: 38057245 DOI: 10.1016/j.ejim.2023.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023]
Affiliation(s)
- James Quinn
- Research Compliance Office, Stanford University, 1705 El Camino Real, MC: 5579, Palo Alto, CA 94306, United States.
| |
Collapse
|