1
|
van Lith TJ, Janssen E, van Dalen JW, Li H, Koeneman M, Sluis WM, Wijers NT, Wermer MJH, Huisman MV, van der Worp HB, Meijer FJA, Tuladhar AM, Bredie SJH, de Leeuw FE. Higher blood pressure variability during hospitalisation is associated with lower cerebral white matter integrity in COVID-19 patients. Blood Press 2025; 34:2493828. [PMID: 40241653 DOI: 10.1080/08037051.2025.2493828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND High blood pressure variability (BPV) is associated with cerebrovascular damage and dementia, but it is unknown whether short-term BPV during hospitalisation is also associated with cerebral white matter (WM) damage. We examined whether BPV, measured in-hospital using continuous monitoring, is associated with WM microstructural integrity in COVID-19 patients. METHODS We included hospitalised COVID-19 patients from the CORONavirus and Ischemic Stroke (CORONIS) study who underwent continuous vital signs monitoring using a wearable device during hospital admission and had an MRI shortly after discharge. Systolic BPV was calculated as Average Real Variability (ARV) and Coefficient of Variation (CV) with 1-, 5- and 20-minute intervals. We used diffusion tensor imaging to assess fractional anisotropy (FA) and peak width of skeletonised mean diffusivity (PSMD) as markers of WM integrity. Associations between BPV and WM integrity were examined with linear regression adjusted for age, mean systolic blood pressure (BP), number of BP measurements and type of respiratory support. RESULTS We included 47 COVID-19 patients (mean age: 59.6 years). BP was measured 6306 ± 4343 times per patient (median admission: 11 days (Interquartile Range [IQR] 7.5-15.0). Both higher ARV and CV were associated with lower WM microstructural integrity, reflected by lower FA (ARV: β = -0.40, p = .010; CV: β = -0.33, p = 0.026) and higher PSMD (CV: β = 0.28, p = .038) after adjustment for confounders. Correction for WM hyperintensities did not change these results. CONCLUSIONS High BPV during hospitalisation is associated with lower WM integrity in COVID-19 patients, although causality needs to be demonstrated. Our findings need validation in hospitalised patients without COVID-19 to examine generalisability.
Collapse
Affiliation(s)
- Theresa J van Lith
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Esther Janssen
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan-Willem van Dalen
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Neurology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Hao Li
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mats Koeneman
- Health Innovation Labs, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wouter M Sluis
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Naomi T Wijers
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke J H Wermer
- Department of Neurology, University Medical Center Groningen, Groningen¸ The Netherlands
| | - Menno V Huisman
- Department of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - H Bart van der Worp
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frederick J A Meijer
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anil M Tuladhar
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sebastian J H Bredie
- Department of Internal Medicine and Health Innovation Labs, Radboudumc, Nijmegen, The Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
2
|
Raju V, Reddy R, Javan AC, Hajihossainlou B, Weissleder R, Guiseppi-Elie A, Kurabayashi K, Jones SA, Faghih RT. Tracking inflammation status for improving patient prognosis: A review of current methods, unmet clinical needs and opportunities. Biotechnol Adv 2025; 82:108592. [PMID: 40324661 PMCID: PMC12173025 DOI: 10.1016/j.biotechadv.2025.108592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/07/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Inflammation is the body's response to infection, trauma or injury and is activated in a coordinated fashion to ensure the restoration of tissue homeostasis and healthy physiology. This process requires communication between stromal cells resident to the tissue compartment and infiltrating immune cells which is dysregulated in disease. Clinical innovations in patient diagnosis and stratification include measures of inflammatory activation that support the assessment of patient prognosis and response to therapy. We propose that (i) the recent advances in fast, dynamic monitoring of inflammatory markers (e.g., cytokines) and (ii) data-dependent theoretical and computational modeling of inflammatory marker dynamics will enable the quantification of the inflammatory response, identification of optimal, disease-specific biomarkers and the design of personalized interventions to improve patient outcomes - multidisciplinary efforts in which biomedical engineers may potentially contribute. To illustrate these ideas, we describe the actions of cytokines, acute phase proteins and hormones in the inflammatory response and discuss their role in local wounds, COVID-19, cancer, autoimmune diseases, neurodegenerative diseases and aging, with a central focus on cardiac surgery. We also discuss the challenges and opportunities involved in tracking and modulating inflammation in clinical settings.
Collapse
Affiliation(s)
- Vidya Raju
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA
| | - Revanth Reddy
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA
| | | | - Behnam Hajihossainlou
- Department of Infectious Diseases, Harlem Medical Center, and Columbia University, New York, 10032, NY, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Department of Systems Biology, Harvard Medical School, and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Anthony Guiseppi-Elie
- Department of Biomedical Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B), and Department of Electrical and Computer Engineering, Texas A & M University, College Station, 77843, TX, USA; Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, Houston, 77030, TX, USA; ABTECH Scientific, Inc., Biotechnology Research Park, Richmond, 23219, Virginia, USA
| | - Katsuo Kurabayashi
- Department of Mechanical and Aerospace Engineering, New York University, New York 11201, NY, USA
| | - Simon A Jones
- Division of Infection and Immunity, and School of Medicine, Cardiff University, UK; Systems Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Rose T Faghih
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA.
| |
Collapse
|
3
|
Alcalá-Santiago Á, Rodriguez-Barranco M, Sánchez MJ, Gil Á, García-Villanova B, Molina-Montes E. Micronutrients, Vitamin D, and Inflammatory Biomarkers in COVID-19: A Systematic Review and Meta-analysis of Causal Inference Studies. Nutr Rev 2025; 83:e1383-e1405. [PMID: 39449666 PMCID: PMC12166185 DOI: 10.1093/nutrit/nuae152] [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] [Indexed: 10/26/2024] Open
Abstract
CONTEXT Experimental and observational studies suggest that circulating micronutrients, including vitamin D (VD), may increase COVID-19 risk and its associated outcomes. Mendelian randomization (MR) studies provide valuable insight into the causal relationship between an exposure and disease outcomes. OBJECTIVES The aim was to conduct a systematic review and meta-analysis of causal inference studies that apply MR approaches to assess the role of these micronutrients, particularly VD, in COVID-19 risk, infection severity, and related inflammatory markers. DATA SOURCES Searches (up to July 2023) were conducted in 4 databases. DATA EXTRACTION AND ANALYSIS The quality of the studies was evaluated based on the MR-STROBE guidelines. Random-effects meta-analyses were conducted where possible. RESULTS There were 28 studies (2 overlapped) including 12 on micronutrients (8 on VD) and COVID-19, 4 on micronutrients (all on VD) and inflammation, and 12 on inflammatory markers and COVID-19. Some of these studies reported significant causal associations between VD or other micronutrients (vitamin C, vitamin B6, iron, zinc, copper, selenium, and magnesium) and COVID-19 outcomes. Associations in terms of causality were also nonsignificant with regard to inflammation-related markers, except for VD levels below 25 nmol/L and C-reactive protein (CRP). Some studies reported causal associations between cytokines, angiotensin-converting enzyme 2 (ACE2), and other inflammatory markers and COVID-19. Pooled MR estimates showed that VD was not significantly associated with COVID-19 outcomes, whereas ACE2 increased COVID-19 risk (MR odds ratio = 1.10; 95% CI: 1.01-1.19) but did not affect hospitalization or severity of the disease. The methodological quality of the studies was high in 13 studies, despite the majority (n = 24) utilizing 2-sample MR and evaluated pleiotropy. CONCLUSION MR studies exhibited diversity in their approaches but do not support a causal link between VD/micronutrients and COVID-19 outcomes. Whether inflammation mediates the VD-COVID-19 relationship remains uncertain, and highlights the need to address this aspect in future MR studies exploring micronutrient associations with COVID-19 outcomes. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42022328224.
Collapse
Affiliation(s)
- Ángela Alcalá-Santiago
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.Granada, 18012 Granada, Spain
- Institute of Nutrition and Food Technology (INYTA) “José Mataix”, Biomedical Research Centre, University of Granada, 18071 Granada, Spain
| | - Miguel Rodriguez-Barranco
- Instituto de Investigación Biosanitaria ibs.Granada, 18012 Granada, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Andalusian School of Public Health, 18012 Granada, Spain
| | - María-José Sánchez
- Instituto de Investigación Biosanitaria ibs.Granada, 18012 Granada, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Andalusian School of Public Health, 18012 Granada, Spain
| | - Ángel Gil
- Instituto de Investigación Biosanitaria ibs.Granada, 18012 Granada, Spain
- Institute of Nutrition and Food Technology (INYTA) “José Mataix”, Biomedical Research Centre, University of Granada, 18071 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
- CIBER de Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Belén García-Villanova
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
| | - Esther Molina-Montes
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.Granada, 18012 Granada, Spain
- Institute of Nutrition and Food Technology (INYTA) “José Mataix”, Biomedical Research Centre, University of Granada, 18071 Granada, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
4
|
Chen M, Nie YK, Liu XY, Liu Y, Guo DY. Antiviral properties of the natural product eugenol: A review. Fitoterapia 2025; 185:106674. [PMID: 40513765 DOI: 10.1016/j.fitote.2025.106674] [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: 03/26/2025] [Revised: 05/20/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Natural small molecule compounds have become the alternative options for developing antiviral reagent due to their broad-spectrum antiviral properties and low toxicity. Eugenol is a natural volatile phenolic aromatic compound primarily derived from clove oil. The small compound is well-recognized for its local anesthetic, anti-inflammatory, and preservative properties, making it a common choice for oral disinfection and as an adjunctive treatment for periodontitis. Clinical and experimental evidence shows that eugenol exhibits significant inhibitory effects against a variety of pathogenic microorganisms, including bacteria, fungi, and the viruses. In viral infectious diseases, eugenol can directly interact with pathogenic antigens in viral structures to prevent viral invasion. Additionally, it offers anti-inflammatory and antioxidant benefits by inhibiting multiple inflammatory mediators, thereby reducing organ damage. To the best of our knowledge, recent advancements in the antiviral properties of eugenol have not been solely addressed. This review aims to offer a new insight into the antiviral activity of eugenol and its potential mechanisms of action. Based on the literature review, we suggest that the antiviral mechanisms of eugenol are associated with cellular autophagy, and that the nuclear factor kappa-B (NF-κB) signaling pathway is the primary inflammatory pathway regulated by eugenol. These findings highlight the need for further investigation through rigorously designed experimental studies focused on specific viruses.
Collapse
Affiliation(s)
- Mei Chen
- Department of Nursing Science, Clinical College of Qilu Medical University, China
| | - Yun-Ke Nie
- Department of Scientific Research, Clinical College of Qilu Medical University, China
| | - Xin-Yu Liu
- Department of Medical education, Clinical College of Qilu Medical University, China
| | - Yang Liu
- Department of Internal Medicine and Pediatric, Clinical College of Qilu Medical University, China; Department of Cardiology, Affiliated Hospital of Qilu Medical University, China.
| | - Dao-Yu Guo
- Department of Neuroscience, Clinical College of Qilu Medical University, China.
| |
Collapse
|
5
|
Zheng Y, He D, Zuo W, Wang W, Wu K, Wu H, Yuan Y, Huang Y, Li H, Lu Y, Zhao L, Wang X, Wang J, Zhang Y, Zou G, Li H, Wang Z, Cao B. Influenza A virus dissemination and infection leads to tissue resident cell injury and dysfunction in viral sepsis. EBioMedicine 2025; 116:105738. [PMID: 40367638 PMCID: PMC12142562 DOI: 10.1016/j.ebiom.2025.105738] [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: 08/20/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Severe respiratory viral infections can lead to viral sepsis (VS), a life-threatening condition characterized by lung and extrapulmonary organ dysfunction. However, the pathology of VS is not clear. Specifically, it is unknown how the cytokine storm and direct virus infection contribute to the damage of extrapulmonary organs. METHODS In this study, we established survival and lethal mouse models of VS by intranasally administering different doses of PR8/H1N1 influenza virus in C57BL/6J male mice, as well as model of bacterial sepsis (BS) caused by Streptococcus pneumoniae as references. Viraemia and extrapulmonary dissemination and infection of the virus were examined. Single-cell sequencing of the lungs and livers was performed at different days post-infection (dpi) in three groups. FINDINGS While bacteria can spread and colonize extensively in extrapulmonary organs, causing multiple organ injuries, IAVs mainly replicate and cause damage in pulmonary cells. Live virus can be isolated in the blood and extrapulmonary organs. Disseminating via the bloodstream, IAVs transiently infect the liver and spleen, causing liver dysfunction and spleen atrophy, without affecting kidney function, despite systematically elevated cytokine levels. Compared to BS, a more significant decrease in the proportion of alveolar macrophages, epithelial cells, endothelial cells, and fibroblasts in the lungs, as well as endothelial cells and Kupffer cells in the liver, was observed in VS. This was accompanied by a longer activated PANoptosis pathway and downregulated genes responsible for barrier function and antigen presentation in the epithelial and endothelial cells. INTERPRETATION Our study suggests that H1N1 influenza virus disseminates through the bloodstream and infects extrapulmonary organs to varying extents, which may lead to differential cell death, organ dysfunction, and trigger VS. FUNDING This research was supported by the National Natural Science Foundation of China (82241056, 82170015, 82030002, 82470007, 824B2001), the National Key R&D Program of China (2023YFC2306300), Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (2021-I2M-1-048), the Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (ZYYCXTD-D-202208), New Cornerstone Science Foundation, National High Level Hospital Clinical Research Funding (2024-NHLHCRF-LX-01-0101, 2024-NHLHCRF-LX-01-0102), Beijing Research Ward Excellence Program (BRWEP2024W114060103), Noncommunicable Chronic Diseases-National Science and Technology Major Project (2023ZD0506200, 2023ZD0506203) and Special Research Fund for Central Universities, Peking Union Medical College (3332024193).
Collapse
Affiliation(s)
- Ying Zheng
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100054, China; National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Di He
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100054, China; National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wenting Zuo
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weiyang Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Kaiwei Wu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hongping Wu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yingying Yuan
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100083, China
| | - Yijiao Huang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, 100084, China
| | - Hongyan Li
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yameng Lu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100054, China
| | - Ling Zhao
- Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiuhong Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jiaying Wang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yulian Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Guming Zou
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Haibo Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; New Cornerstone Science Laboratory, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Zai Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100083, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100054, China; National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, 100084, China; New Cornerstone Science Laboratory, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
6
|
Küng AJ, Dykun I, Totzeck M, Mincu R, Michel L, Kill C, Witzke O, Buer J, Rassaf T, Mahabadi AA. Epicardial adipose tissue in patients with and without COVID-19 infection. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2025; 54:100548. [PMID: 40322277 PMCID: PMC12049814 DOI: 10.1016/j.ahjo.2025.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 02/09/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025]
Abstract
Background Acute COVID-19 infection frequently affects the cardiovascular system and causes acute myocardial injury. Epicardial Adipose Tissue (EAT), a visceral adipose tissue surrounding the myocardium and coronary arteries, has unique paracrine and endocrine effects, modulating the heart's inflammatory environment. Systemic inflammation stimulates TNF-α and Interleukin-6 secretion from EAT, contributing to cytokine storms and intensifying systemic responses. We aimed to determine whether EAT amount differs in patients with and without acute COVID-19 infection and myocardial injury. Methods This study analyzed the CoV-COR registry cohort, conducted at the University Hospital Essen, including patients with symptoms suggestive of COVID-19 infection. The infection was confirmed by PCR. EAT thickness was measured by two-dimensional TTE. Results A total of 296 patients (mean age 63.6 ± 17.26 years, 55.4 % male) were included. Patients with confirmed COVID-19 infection were younger, more frequently treated with antihypertensive medication, and had higher BMI and systolic blood pressures. Univariate logistic regression showed no association between EAT and myocardial injury 0.97 (0.74; 1.28, p = 0.82). A trend towards an association was observed between increasing EAT thickness and COVID-19 infection 1.25 (0.99; 1.59, p = 0.060). Adjusting for age and gender strengthened the association, with a 48 % (1.14; 1.93, p = 0.004) increased odds of COVID-19 infection per increase in EAT thickness. Multivariable regression yielded consistent effect sizes 1.47 (1.01; 2.16, p = 0.047). Conclusion EAT thickness is associated with the presence of an acute COVID-19 infection but not with a myocardial injury. Further research is needed to assess if systemic viral infection induces dynamic changes in EAT.
Collapse
Affiliation(s)
- Alexander J. Küng
- West German Heart and Vascular Center Essen, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstr, 55, 45147 Essen, Germany
| | - Iryna Dykun
- West German Heart and Vascular Center Essen, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstr, 55, 45147 Essen, Germany
| | - Matthias Totzeck
- West German Heart and Vascular Center Essen, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstr, 55, 45147 Essen, Germany
| | - Raluca Mincu
- West German Heart and Vascular Center Essen, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstr, 55, 45147 Essen, Germany
| | - Lars Michel
- West German Heart and Vascular Center Essen, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstr, 55, 45147 Essen, Germany
| | - Clemens Kill
- Center for Emergency Medicine, University Hospital Essen, Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, Germany
| | - Tienush Rassaf
- West German Heart and Vascular Center Essen, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstr, 55, 45147 Essen, Germany
| | - Amir A. Mahabadi
- West German Heart and Vascular Center Essen, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstr, 55, 45147 Essen, Germany
| |
Collapse
|
7
|
Chen J, Zhao S, Yan H, Huang Y, Wei C, Liu J, Sun J. Plasma SARS-CoV-2 nucleocapsid antigen levels are associated with lung infection and tissue-damage biomarkers. Virus Res 2025; 356:199580. [PMID: 40339608 DOI: 10.1016/j.virusres.2025.199580] [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: 01/11/2025] [Revised: 04/17/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND SARS-CoV-2 nucleocapsid (N) antigen has been confirmed in the peripheral blood of patients with new coronavirus infection,yet its diagnostic and prognostic significance remains unclear. This study aimed to characterize the dynamics of SARS-CoV-2 N antigenemia in patients with novel coronavirus positivity, and to assess its potential association with clinical severity and plasma biomarker levels. METHODS We analyzed the level of SARS-CoV-2 N antigen, spike receptor-binding domain (S-RBD) IgG, neutralizing antibodies (NAb) and tissue-damage biomarkers was assessed in 180 plasma samples from 51 SARS-CoV-2-positive individuals. Plasma antigen levels were compared with concurrent respiratory nucleic acid amplification test results. RESULTS Patients with Ct values below 30 showed significantly different serum antigen levels compared to those with Ct values above 30 (p < 0.01). However, no significant positive correlation was found between respiratory viral load and serum antigen levels. Further analysis revealed that patients with pneumonia had markedly higher serum antigen levels than those without (p < 0.0001). Additionally, serum amyloid A (SAA) and ferritin (Fe) levels were significantly elevated in the antigenemia-positive group compared to the negative group, while procalcitonin (PCT) and interleukin-6 (IL-6) levels showed no significant differences. Notably, the positivity rate of N antigen in peripheral blood peaked at 47.1% (95% CI: 37.8%-56.7%) during the first week of infection and then gradually decreased over time. Moreover, patients with severe COVID-19 exhibited significantly higher serum antigen levels than those with mild or moderate disease (p < 0.0001). Serum levels of SARS-CoV-2 S-RBD IgG and neutralizing antibodies (NAb) were also significantly higher in antigenemia-negative patients than in antigenemia-positive patients (p < 0.0001). CONCLUSIONS Our findings highlight the multifaceted role of antigenemia in SARS-CoV-2 and suggest its potential as a biomarker for disease monitoring and risk stratification.
Collapse
Affiliation(s)
- Jing Chen
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Shuai Zhao
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Haiyang Yan
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Yaomeng Huang
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Congzhen Wei
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Jiajia Liu
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China.
| | - Jingna Sun
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China.
| |
Collapse
|
8
|
Huynh DTN, Nguyen HT, Hsieh CM. Taiwan Chingguan Yihau may improve post-COVID-19 respiratory complications through PI3K/AKT, HIF-1, and TNF signaling pathways revealed by network pharmacology analysis. Mol Divers 2025; 29:2305-2321. [PMID: 39382736 DOI: 10.1007/s11030-024-10993-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024]
Abstract
The emergence of new SARS-CoV-2 variants with a higher contagious capability and faster transmissible speed has imposed an incessant menace on global health and the economy. The SARS-CoV-2 infection might reoccur and last much longer than expected. Thence, there is a high possibility that COVID-19 can cause long-term health problems. This condition needs to be investigated thoroughly, especially the post-COVID-19 complications. Respiratory tract disorders are common and typical complications after recovery. Until now, there has been a lack of data on specialized therapeutic medicine for post-COVID-19 complications. The clinical efficacy of NRICM101 has been demonstrated in hospitalized COVID-19 patients. This herbal medicine may also be a promising therapy for post-COVID-19 complications, thanks to its phytochemical constituents. The potential pharmacological mechanisms of NRICM101 in treating post-COVID-19 respiratory complications were investigated using network pharmacology combined with molecular docking, and the results revealed that NRICM101 may exert a beneficial effect through the three primary pathways: PI3K/AKT, HIF-1, and TNF signaling pathways. Flavonoids (especially quercetin) have a predominant role and synergize with other active compounds to produce therapeutic effectiveness. Most of the main active compounds exist in three chief herbal ingredients, including Liquorice root (Glycyrrhiza glabra), Scutellaria root (Scutellaria baicalensis), and Mulberry leaf (Morus alba). To our knowledge, this is the first study of the NRICM101 effect on post-COVID-19 respiratory complications. Our findings may provide a better understanding of the potential mechanisms of NRICM101 in treating SARS-CoV-2 infection and regulating the immunoinflammatory response to improve post-COVID-19 respiratory complications.
Collapse
Affiliation(s)
- Dung Tam Nguyen Huynh
- College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Can Tho University of Medicine and Pharmacy, Can Tho City, 94117, Vietnam
| | - Hien Thi Nguyen
- Department of Nutrition and Food Safety, Faculty of Public Health, Can Tho University of Medicine and Pharmacy, Can Tho City, 94117, Vietnam
| | - Chien-Ming Hsieh
- College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
9
|
Feira MF, Sbruzzi RC, Maciel-Fiuza MF, Griebeler VC, Gregianini TS, Martins LG, Cadore NA, Chies JAB, Kowalski TW, Vianna FSL. Association Between Genetic Variants in TNF, IL6, and IL1B Genes and Severity of COVID-19: A Cross-Sectional Study of Patients from Southern Brazil. Diagnostics (Basel) 2025; 15:1403. [PMID: 40506975 PMCID: PMC12154427 DOI: 10.3390/diagnostics15111403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/18/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025] Open
Abstract
Background/Objectives: Genetic variants in cytokine genes such as IL1B, IL6, and TNF may influence inflammatory responses to SARS-CoV-2 and affect disease severity. This study investigates the role of these variants in relation to COVID-19 outcomes, including hospitalization, ICU admission, and mortality. Methods: A total of 500 unvaccinated individuals from southern Brazil diagnosed with COVID-19 via RT-PCR were analyzed. DNA was extracted from nasopharyngeal swabs and genotyped for functional variants selected based on evidence of regulatory function and prior associations with inflammatory outcomes-IL1B (rs4848306, rs1143623, rs16944, rs1143627), IL6 (rs1800795, rs2069832, rs2069840, rs2069845), and TNF (rs1799964, rs1800630, rs1799724, rs1800629, rs361525). Multivariate logistic regression analysis, adjusted for sex and age, was employed to assess the association between these genetic variants and severe clinical outcomes. Results: The results indicated that the IL1B rs16944-AG (OR: 1.98 [95% CI: 1.22-3.23], p = 0.006) and TNF rs1799964-CT (OR: 1.97 [95% CI: 1.22-3.22], p = 0.006) genotypes were associated with the need for hospitalization, while TNF rs1800630-AA (OR: 2.37 [95% CI: 1.08-5.33], p = 0.034) was associated with ICU admission. Additionally, the CC genotype of TNF rs1799964 was associated with a higher risk of mortality (OR: 3.73 [95% CI: 1.21-14.37], p = 0.034). Conclusions: Genetic variants-specifically IL1B rs16944 and rs1143627, and TNF rs1799964 and rs1800630-were associated with COVID-19 severity and should be further investigated in larger studies to evaluate their potential as predictive markers of severe outcomes in COVID-19.
Collapse
Affiliation(s)
- Mariléa Furtado Feira
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil; (M.F.F.); (R.C.S.); (M.F.M.-F.); (N.A.C.); (J.A.B.C.); (T.W.K.)
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil;
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Renan Cesar Sbruzzi
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil; (M.F.F.); (R.C.S.); (M.F.M.-F.); (N.A.C.); (J.A.B.C.); (T.W.K.)
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil;
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Miriãn Ferrão Maciel-Fiuza
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil; (M.F.F.); (R.C.S.); (M.F.M.-F.); (N.A.C.); (J.A.B.C.); (T.W.K.)
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil;
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Vitória Carolina Griebeler
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil;
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Tatiana Schaffer Gregianini
- Laboratório Central do Centro Estadual de Vigilância em Saúde da do Rio Grande do Sul (LACEN), Secretaria da Saúde do estado do Rio Grande do Sul (LACEN/CEVS/SES-RS), Porto Alegre 90620-000, RS, Brazil;
| | - Letícia Garay Martins
- Centro Estadual de Vigilância em Saúde da Secretaria de Saúde do estado do Rio Grande do Sul (CEVS/SES-RS), Porto Alegre 90620-000, RS, Brazil;
| | - Nathan Araujo Cadore
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil; (M.F.F.); (R.C.S.); (M.F.M.-F.); (N.A.C.); (J.A.B.C.); (T.W.K.)
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil;
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Jose Artur Bogo Chies
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil; (M.F.F.); (R.C.S.); (M.F.M.-F.); (N.A.C.); (J.A.B.C.); (T.W.K.)
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Thayne Woycinck Kowalski
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil; (M.F.F.); (R.C.S.); (M.F.M.-F.); (N.A.C.); (J.A.B.C.); (T.W.K.)
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil;
- Laboratory Genetics Unit, Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil
| | - Fernanda Sales Luiz Vianna
- Postgraduate Program in Genetics and Molecular Biology (PPGBM), Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil; (M.F.F.); (R.C.S.); (M.F.M.-F.); (N.A.C.); (J.A.B.C.); (T.W.K.)
- Laboratory of Genomic Medicine, Center of Experimental Research, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil;
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| |
Collapse
|
10
|
Sancho-Saldaña A, Gil-Sánchez A, Quirant-Sánchez B, Boigues M, Canudes M, Peralta S, Solana MJ, González-Mingot C, Quibus L, Martínez-Cáceres E, Torres P, Hervás JV, Moreno-Magallon J, Brieva L. Profile of Cytokines Associated with SARS-CoV2 Seropositivity in Multiple Sclerosis Patients and Its Persistence over Six Months. J Clin Med 2025; 14:3736. [PMID: 40507498 PMCID: PMC12155705 DOI: 10.3390/jcm14113736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/08/2025] [Accepted: 05/18/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Patients with multiple sclerosis (pwMS) receiving disease-modifying therapies (DMTs) may exhibit altered immune responses to infections such as SARS-CoV-2. This study aimed to characterize the cytokine profiles associated with prior SARS-CoV-2 infection and to identify immune markers related to the persistence of the humoral response in pwMS. Methods: A total of 90 pwMS were recruited before the introduction of COVID-19 vaccination in Spain; 46 were seropositive-defined by the presence of IgG, IgM, or IgA antibodies against SARS-CoV-2-and 44 were seronegative. We compared baseline cytokine levels between groups and followed seropositive individuals for six months to assess IgG antibody persistence. Results: Seropositive patients showed significantly lower baseline levels of IL-10, IL-23, and IFN-α compared to seronegative individuals. Notably, elevated IL-18 at baseline was associated with persistent IgG seropositivity at six months. Conclusions: These findings suggest a distinct cytokine profile in SARS-CoV-2-exposed pwMS and highlight IL-18 as a potential marker of sustained humoral response. This study provides insight into host-virus immune dynamics in MS patients and may help guide future strategies for infection monitoring and immune evaluation in this population.
Collapse
Affiliation(s)
- Agustín Sancho-Saldaña
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Anna Gil-Sánchez
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Bibiana Quirant-Sánchez
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
- Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Cerdanyola del Valles, Spain
| | - Marc Boigues
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
| | - Marc Canudes
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Silvia Peralta
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - María José Solana
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Cristina González-Mingot
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Department of Medicine, University of Lleida (UdL), 25198 Lleida, Spain
| | - Laura Quibus
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Eva Martínez-Cáceres
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
- Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Cerdanyola del Valles, Spain
| | - Pascual Torres
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - José Vicente Hervás
- Servicio de Neurología, Hospital de Sant Joan Despí Moisés Broggi, 08970 Barcelona, Spain;
| | - Judith Moreno-Magallon
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Luis Brieva
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Department of Medicine, University of Lleida (UdL), 25198 Lleida, Spain
| |
Collapse
|
11
|
Hingole P, Saha P, Das S, Gundu C, Kumar A. Exploring the role of mitochondrial dysfunction and aging in COVID-19-Related neurological complications. Mol Biol Rep 2025; 52:479. [PMID: 40397294 DOI: 10.1007/s11033-025-10586-0] [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: 03/27/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025]
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, posed a tremendous challenge to healthcare systems globally. Severe COVID-19 infection was reported to be associated with altered immunometabolism and cytokine storms, contributing to poor clinical outcomes and in many cases resulting in mortality. Despite promising preclinical results, many drugs have failed to show efficacy in clinical trials, highlighting the need for novel approaches to combat the virus and its severe manifestations. Mitochondria, crucial for aerobic respiration, play a pivotal role in modulating immunometabolism and neuronal function, making their compromised capability as central pathological mechanism contributing to the development of neurological complications in COVID-19. Dysregulated mitochondrial dynamics can lead to uncontrolled immune responses, underscoring the importance of mitochondrial regulation in shaping clinical outcomes. Aging further accelerates mitochondrial dysfunction, compounding immune dysregulation and neurodegeneration, making older adults particularly vulnerable to severe COVID-19 and its neurological sequelae. COVID-19 infection impairs mitochondrial oxidative phosphorylation, contributing to the long-term neurological complications associated with the disease. Additionally, recent reports also suggest that up to 30% of COVID-19 patients experience lingering neurological issues, thereby highlighting the critical need for further research into mitochondrial pathways to mitigate long-tern neurological consequences of Covid-19. This review examines the role of mitochondrial dysfunction in COVID-19-induced neurological complications, its connection to aging, and potential biomarkers for clinical diagnostics. It also discusses therapeutic strategies aimed at maintaining mitochondrial integrity to improve COVID-19 outcomes.
Collapse
Affiliation(s)
- Prajakta Hingole
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, 168, Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Priya Saha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) SAS Nagar, Sec 67, Mohali, 160062, Punjab, India
| | - Sourav Das
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) SAS Nagar, Sec 67, Mohali, 160062, Punjab, India
| | - Chayanika Gundu
- Department of Ophthalmology, University of Wisconsin, Madison, USA
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, 168, Maniktala Main Road, Kolkata, 700054, West Bengal, India.
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) SAS Nagar, Sec 67, Mohali, 160062, Punjab, India.
| |
Collapse
|
12
|
Ortega Á, Duran P, Garrido B, Manzano A, Navarro C, Silva A, Rojas M, De Sanctis JB, Radzioch D, Rivera-Porras D, Paredes CS, Bermúdez V. Specialized Pro-Resolving Lipid Mediators in Pulmonary Diseases: Molecular and Therapeutic Implications. Molecules 2025; 30:2212. [PMID: 40430385 PMCID: PMC12114278 DOI: 10.3390/molecules30102212] [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: 03/04/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Inflammatory lung diseases (ILDs) represent a global public health crisis characterized by escalating prevalence, significant morbidity, and substantial mortality. In response to the complex immunopathogenic mechanisms driving these conditions, novel pharmacological strategies targeting resolution pathways have emerged throughout the discovery of specialized pro-resolving lipid mediator (SPM; resolvins, maresins, and protectins) dysregulation across the ILD spectra, positioning these endogenous molecules as promising therapeutic candidates for modulating maladaptive inflammation and promoting tissue repair. Over the past decade, this paradigm has catalyzed extensive translational research into SPM-based interventions as precision therapeutics for respiratory inflammation. In asthma, they reduce mucus hypersecretion, bronchial hyperreactivity, and airway inflammation, with prenatal SPM exposure potentially lowering offspring disease risk. In COPD, SPMs attenuate amyloid A-driven inflammation, normalizing cytokine/chemokine imbalances and oxidative stress and mitigating COVID-19-associated cytokine storm, enhancing survival. This review synthesizes SPMs' pharmacotherapeutic mechanisms in ILDs and evaluates current preclinical and clinical evidence.
Collapse
Affiliation(s)
- Ángel Ortega
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4001, Venezuela; (Á.O.); (P.D.); (B.G.); (A.M.); (C.N.); (A.S.); (M.R.)
| | - Pablo Duran
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4001, Venezuela; (Á.O.); (P.D.); (B.G.); (A.M.); (C.N.); (A.S.); (M.R.)
| | - Bermary Garrido
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4001, Venezuela; (Á.O.); (P.D.); (B.G.); (A.M.); (C.N.); (A.S.); (M.R.)
| | - Alexander Manzano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4001, Venezuela; (Á.O.); (P.D.); (B.G.); (A.M.); (C.N.); (A.S.); (M.R.)
| | - Carolina Navarro
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4001, Venezuela; (Á.O.); (P.D.); (B.G.); (A.M.); (C.N.); (A.S.); (M.R.)
| | - Aljadis Silva
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4001, Venezuela; (Á.O.); (P.D.); (B.G.); (A.M.); (C.N.); (A.S.); (M.R.)
| | - Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4001, Venezuela; (Á.O.); (P.D.); (B.G.); (A.M.); (C.N.); (A.S.); (M.R.)
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, 77900 Olomouc, Czech Republic;
| | - Danuta Radzioch
- The Research Institute of the McGill, University Health Center, McGill University, Montreal, QC H0H H9Z, Canada;
| | - Diego Rivera-Porras
- Universidad de la Costa, Departamento de Productividad e Innovación, Barranquilla 080001, Atlántico, Colombia;
| | - Carlos Silva Paredes
- Universidad del Zulia, Facultad de Medicina, Departamento de Ciencias Fisiológicas, Maracaibo 4001, Venezuela;
| | - Valmore Bermúdez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Centro de Investigaciones en Ciencias de la Vida, Barranquilla 080001, Atlántico, Colombia
| |
Collapse
|
13
|
Pius-Sadowska E, Kulig P, Niedźwiedź A, Baumert B, Rogińska D, Łuczkowska K, Sobuś A, Parczewski M, Kawa M, Paczkowska E, Machaliński B. The micro-RNA expression profile predicts the severity of SARS-CoV-2 infection. Sci Rep 2025; 15:17139. [PMID: 40382351 DOI: 10.1038/s41598-025-01229-2] [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: 10/23/2024] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Abstract
Although much is known about the pathophysiology of severe COVID-19, there are still areas that remain to be determined. It is well established that the pivotal molecular event is a hyperinflammatory response also referred to as a cytokine storm. The aim of this retrospective cohort study was to determine miRNAs which might be predictive for the admission to the intensive care unit (ICU). We analyzed blood samples from 210 COVID-19 patients and the control group consisted of 80 healthy individuals. Results revealed the miRNA expression pattern has the potential to predict the severity of COVID-19, reflecting the clinical symptoms of the infection, such as the need for oxygen therapy and concomitant pneumonia. In particular, low expression of miRNAs miR106a-5p, miR17-5p, miR181a-5p, miR191-5p, miR20a-5p and miR451a, especially in the initial phase of the disease, is associated with an unfavorable clinical course of SARS-CoV-2 infection (admission to the ICU).
Collapse
Affiliation(s)
- Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Piotr Kulig
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Anna Niedźwiedź
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Bartłomiej Baumert
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Dorota Rogińska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Anna Sobuś
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, Arkońska 4 Street, 71-455, Szczecin, Poland
| | - Miłosz Kawa
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland.
| |
Collapse
|
14
|
Yao Z, Feng Z, Zhang H, Zhang B. ScRNA-Seq reveals T cell immunity in COVID-19 patients and implications for immunotherapy. Int Immunopharmacol 2025; 155:114663. [PMID: 40233451 DOI: 10.1016/j.intimp.2025.114663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
SARS-CoV-2, the virus causing COVID-19, poses significant health threats due to its high transmissibility and potential for severe respiratory complications. T cells, central to adaptive immunity, also interact with innate immunity, playing a pivotal role in coordinating defenses and eliminating infected cells. Single-cell RNA sequencing (scRNA-seq) has provided more subtle heterogeneity, rare subpopulations, or new subpopulations that are at the district differentiation stage or with specific function. Thus, elucidating how T cell heterogeneity impacts COVID-19 disease severity remains a critical question requiring comprehensive analysis. This review revealed the heterogeneity of the host T cells, including conventional T cells (CD8+, CD4+ T cells) and unconventional T cells, including natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) and gamma-delta T (γδT) cells in COVID-19 patients with different clinical manifestations. Severe COVID-19 had marked lymphopenia, excessive activation, elevated exhaustion and reduced functional diversity of T cells. Pathogenic contributions arise from dysregulated cytotoxic T cells, Treg cells and unconventional T cells collectively driving systemic hyperinflammation and tissue injury. Current therapeutic strategies targeting T cells-such as enhancing virus-specific T cell responses, reverting T-cell exhaustion and alleviating inflammation-exhibit inconsistent efficacy, underscoring the need for combinatorial approaches. This review highlights how scRNA-seq deciphers T cell heterogeneity and dysfunction in COVID-19. By targeting T cell exhaustion, inflammation, and subset-specific deficits, these insights pave the way for therapies and vaccines.
Collapse
Affiliation(s)
- Zhihong Yao
- Faculty of Clinical Medicine, Hanzhong Vocational and Technical College, Hanzhong 723002, China; Affiliated Hospital, Hanzhong Vocational and Technical College, Hanzhong 723012, China; Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhao Feng
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Hui Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, China.
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
15
|
Tian L, Zhao Z, Gao W, Liu Z, Li X, Zhang W, Li Z. SARS-CoV-2 nsp16 is regulated by host E3 ubiquitin ligases, UBR5 and MARCHF7. eLife 2025; 13:RP102277. [PMID: 40358464 PMCID: PMC12074641 DOI: 10.7554/elife.102277] [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] [Indexed: 05/15/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), remains a global public health threat with considerable economic consequences. The nonstructural protein 16 (nsp16), in complex with nsp10, facilitates the final viral mRNA capping step through its 2'-O-methylase activity, helping the virus to evade host immunity and prevent mRNA degradation. However, nsp16 regulation by host factors remains poorly understood. While various E3 ubiquitin ligases interact with SARS-CoV-2 proteins, their roles in targeting nsp16 for degradation remain unclear. In this study, we demonstrate that nsp16 undergoes ubiquitination and proteasomal degradation mediated by the host E3 ligases UBR5 and MARCHF7. UBR5 induces K48-linked ubiquitination, whereas MARCHF7 promotes K27-linked ubiquitination, independently suppressing SARS-CoV-2 replication in cell cultures and in mice. Notably, UBR5 and MARCHF7 also degrade nsp16 variants from different viral strains, exhibiting broad-spectrum antiviral activity. Our findings reveal novel antiviral mechanisms of the ubiquitin-proteasome system (UPS) and highlight their potential therapeutic targets against COVID-19.
Collapse
Affiliation(s)
- Li Tian
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zongzheng Zhao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Wenying Gao
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zirui Liu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Xiao Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Wenyan Zhang
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zhaolong Li
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
16
|
Liu S, Xu W, Tu B, Xiao Z, Li X, Huang L, Yuan X, Zhou J, Yang X, Yang J, Chang D, Chen W, Wang FS. Early Immunological and Inflammation Proteomic Changes in Elderly COVID-19 Patients Predict Severe Disease Progression. Biomedicines 2025; 13:1162. [PMID: 40426989 PMCID: PMC12108611 DOI: 10.3390/biomedicines13051162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Elderly patients infected with SARS-CoV-2 are at higher risk of developing cytokine storm and severe outcomes; however, specific immunological and proteomic biomarkers for early prediction remain unclear in this vulnerable group. Methods: We enrolled 182 elderly COVID-19 patients from the Chinese PLA General Hospital between November 2022 and April 2023, categorizing them based on progression to respiratory failure requiring mechanical ventilation (defined as severe progression). Olink proteomic analysis was performed on admission serum from 40 propensity score-matched samples, with differentially expressed proteins (DEPs) validated by cytometric bead array (CBA) in 178 patients. To predict severe progression, a model was developed using a 70% training set and validated on a 30% validation set. LASSO regression screened features followed by logistic regression and receiver operating characteristic (ROC) analysis to optimize the model by incrementally incorporating features ranked by random forest importance. Results: Elderly patients progressing to severe COVID-19 exhibited early immune dysregulation, including neutrophilia, lymphopenia, monocytopenia, elevated procalcitonin (PCT), C-reactive protein (CRP), interleukin-6 (IL-6), neutrophil-to-lymphocyte ratio (NLR), and systemic immune-inflammation index (SII), as well as coagulation dysfunction and multi-organ injury. Proteomics identified a set of biomarkers, including tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and revealed disruptions in signaling pathways, including the mTOR and VEGF signaling pathways. The optimal predictive model, which incorporated PCT, IL-6, monocyte percentage, lymphocyte count, and TRAIL, achieved an area under curve (AUC) of 0.870 (0.729-1.000) during validation. TRAIL levels negatively correlated with fibrinogen (p < 0.05). Conclusions: Elderly COVID-19 patients with severe progression demonstrate early immune dysregulation, hyperinflammation, coagulation dysfunction, and multi-organ injury. The model we proposed effectively predicts disease progression in elderly COVID-19 patients, providing potential biomarkers for early clinical risk stratification in this vulnerable population.
Collapse
Affiliation(s)
- Shiyang Liu
- Medical School of Chinese PLA, Beijing 100853, China;
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Wen Xu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Bo Tu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Zhiqing Xiao
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100007, China; (Z.X.); (D.C.)
| | - Xue Li
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
- Yu-Yue Pathology Scientific Research Center, Chongqing 401329, China
| | - Lei Huang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Xin Yuan
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Juanjuan Zhou
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Xinxin Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Junlian Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - De Chang
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100007, China; (Z.X.); (D.C.)
| | - Weiwei Chen
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| |
Collapse
|
17
|
Vanderkamp SG, Niazy M, Stegelmeier AA, Stinson KJ, Ricker N, Bridle BW. Cytokine, chemokine, and acute-phase protein profiles in plasma as correlative biomarkers of clinical outcomes for patients with COVID-19. Sci Rep 2025; 15:15397. [PMID: 40316702 PMCID: PMC12048561 DOI: 10.1038/s41598-025-99248-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/17/2025] [Indexed: 05/04/2025] Open
Abstract
Coronavirus disease identified in 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus-2, had a global impact on human health and the economy. The aim of this study was to quantify cytokines, chemokines, and acute phase proteins in the plasma of patients with COVID-19 to elucidate potential biomarkers to inform prognostic and treatment decisions. Clustering analysis using the K-prototypes method identified underlying biological patterns in patients with COVID-19. The penalized multinomial logistic regression analysis identified two comorbidities (hypertension, congestive heart failure) and thirteen analytes as potential risk factors for COVID-19 progression with 88.2% accuracy. Based on a patient's age, high concentrations of interleukin (IL)-6, monocyte chemoattractant protein-1, and pentraxin 3 were important biomarkers for lethal COVID-19. Decreased concentrations of interferon gamma-induced protein-10, IL-10, and soluble tumor necrosis factor receptor I were found to be associated with mild COVID-19, while increasing concentrations of these analytes could be used to predict COVID-19 severity.
Collapse
Affiliation(s)
- Sierra G Vanderkamp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Maysa Niazy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | | | - Nicole Ricker
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
18
|
Bagriacik U, Karakus R, Yaman M, Oruklu N, Araz M, Kalayci Z, Ozger HS, Yildiz Y, Senol E. Increased serum levels of IL-40 are associated with IgA and NETosis biomarkers in Covid-19 patients: IL-40 and infectious diseases. PLoS One 2025; 20:e0321578. [PMID: 40315230 PMCID: PMC12047749 DOI: 10.1371/journal.pone.0321578] [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: 10/04/2024] [Accepted: 03/07/2025] [Indexed: 05/04/2025] Open
Abstract
Interleukin 40 (IL-40) is a novel cytokine that has been associated with B lymphocytes, particularly IgA-secreting cells in gut mucosa. Considering mucosal association of IL-40, we aimed to determine serum IL-40 levels in patients with Covid-19. We compared IL-40 concentrations in healthy people to both patients with mild symptoms of SARS-CoV-2 infection and pneumonia. IL-40 was measured by ELISA. Serum IgA levels were tested by nephelometry. For the first time, we demonstrated that SARS-CoV-2 infection increased serum IL-40 levels significantly. The elevation of IL-40 in serum was related to severity of the infection. Therefore, IL-40 concentrations were quite higher in patients with symptoms of pneumonia. Our findings indicated that IgA and NETosis biomarkers were related with IL-40 increase. Based on these findings, we speculated that IL-40 would be associated with immune activities in the mucosa of the lungs of SARS-CoV-2-infected patients. This association may be linked to a mechanism that has a control on IgA and NETosis.
Collapse
Affiliation(s)
- Umit Bagriacik
- Department of Immunology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| | - Resul Karakus
- Department of Immunology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| | - Melek Yaman
- Department of Immunology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| | - Nihan Oruklu
- Department of Immunology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| | - Milat Araz
- Department of Immunology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| | - Zafer Kalayci
- Department of Immunology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| | - Hasan Selcuk Ozger
- Department of Infectious Diseases and Clinical Microbiology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| | - Yesim Yildiz
- Department of Infectious Diseases and Clinical Microbiology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| | - Esin Senol
- Department of Infectious Diseases and Clinical Microbiology, Gazi University, Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
19
|
Aghadoost S, Molazeinal Y, Khoddami SM, Shokuhifar G, Dabirmoghaddam P, Saffari M. Dysphonia Severity Index and Consensus Auditory-Perceptual Evaluation of Voice Outcomes, and Their Relation in Hospitalized Patients with COVID-19. J Voice 2025; 39:853.e1-853.e8. [PMID: 36642593 PMCID: PMC9712076 DOI: 10.1016/j.jvoice.2022.11.034] [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: 09/22/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
OBJECTIVES This study aimed to compare the results of the Dysphonia Severity Index (DSI) and Consensus Auditory-Perceptual Evaluation of Voice (CAPE-V) between patients hospitalized with COVID-19 and healthy subjects, as well as to investigate the correlation between DSI and CAPE-V. STUDY DESIGN Cross-sectional survey. MATERIAL AND METHODS Eighty subjects, 40 COVID-19 patients (with a mean age of 41.2± 5.41) and 40 healthy subjects (with a mean age of 44.50± 3.50) participated in this study. Assessments included the DSI for aerodynamic-acoustic measurement and the Persian version of Consensus Auditory-Perceptual Evaluation of Voice (CAPE-V) for evaluating auditory-perceptual voice quality. Data were analyzed by means of the independent t-test and Pearson correlation at the 5% significance level. RESULTS The results showed COVID-19 patients got significantly lower score in DSI compared to healthy subjects (P < 0.05). Moreover, the patients with COVID-19 had higher scores in all categories of voice production (severity, roughness, loudness, pitch, strain and breathiness) than the healthy group (P < 0.05). Comparing the result of the two voice assessments in each group revealed that there was a greater negative significant correlation in the diseased group (r p: -0.68, P: 0.001) than in the healthy group (r p: -0.37,P: 0.049). CONCLUSIONS Hospitalized COVID-19 patients experience deviations in the voice quality and acoustic-aerodynamic features of their voice. Also, the results of this study showed the patient group had higher perceptual dysphonia and lower voice quality compared to the healthy group. Further studies are recommended to determine the relationship between objective and subjective voice evaluation in patients with COVID-19 after recovery.
Collapse
Affiliation(s)
- Samira Aghadoost
- Department of Speech Therapy, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Tehran, Iran.
| | - Yasamin Molazeinal
- School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Tehran, Iran
| | - Seyyedeh Maryam Khoddami
- Department of Speech Therapy, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Tehran, Iran
| | - Ghazaal Shokuhifar
- Department of audiology, University of Social Welfare and Rehabilitation, Tehran, Tehran, Iran
| | - Payman Dabirmoghaddam
- Otolaryngology Research Center, Tehran University of Medical Sciences, Tehran, Tehran, Iran
| | - Maryam Saffari
- Department of radiology, faculty of medicine, Kashan University of Medical Sciences, Tehran, Tehran, Iran
| |
Collapse
|
20
|
David C, Verney C, Si-Tahar M, Guillon A. Evaluating the evidence for GM-CSF as a host-directed therapy in respiratory infections. Cytokine 2025; 189:156902. [PMID: 39999678 DOI: 10.1016/j.cyto.2025.156902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
Novel therapeutic approaches are needed to treat respiratory infections due to the rising antimicrobial resistance and the lack of effective antiviral therapies. A promising avenue to overcome treatment failure is to develop strategies that target the host immune response rather than the pathogen itself. Granulocyte-macrophage colony-stimulating factor (GM-CSF) plays a critical role in controlling homeostasis in lungs, alveolar macrophages being the most sensitive cells to GM-CSF signaling. In this review, we discuss the importance of GM-CSF secretion for lung homeostasis and its alteration during respiratory infections. We also present the pre-clinical evidence and clinical investigations evaluating GM-CSF-based treatments (administration or inhibition) as a therapeutic strategy for treating respiratory infections, highlighting both supporting and contradictory findings.
Collapse
Affiliation(s)
- Camille David
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France
| | - Charles Verney
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France
| | - Antoine Guillon
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France; CHRU de Tours, Service de Médecine Intensive Réanimation, Tours, France.
| |
Collapse
|
21
|
Wang J, Xiong Y, Song Z, Li Y, Zhang L, Qin C. Progress in research on osteoporosis secondary to SARS-CoV-2 infection. Animal Model Exp Med 2025; 8:829-841. [PMID: 40029778 DOI: 10.1002/ame2.12573] [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: 09/12/2024] [Accepted: 01/13/2025] [Indexed: 05/28/2025] Open
Abstract
The World Health Organization has declared that COVID-19 no longer constitutes a "public health emergency of international concern," yet the long-term impact of SARS-CoV-2 infection on bone health continues to pose new challenges for global public health. In recent years, numerous animal model and clinical studies have revealed that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can lead to secondary osteoporosis. The mechanisms involved are related to the virus's direct effects on bone tissue, dysregulation of the body's inflammatory response, hypoxia, noncoding RNA imbalance, and metabolic abnormalities. Although these studies have unveiled the connection between SARS-CoV-2 infection and osteoporosis, current research is not comprehensive and in depth. Future studies are needed to evaluate the long-term effects of SARS-CoV-2 on bone density and metabolism, elucidate the specific mechanisms of pathogenesis, and explore potential interventions. This review aims to collate existing research literature on SARS-CoV-2 infection-induced secondary osteoporosis, summarize the underlying mechanisms, and provide direction for future research.
Collapse
Affiliation(s)
- Jinlong Wang
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
- Changping National Laboratory (CPNL), Beijing, China
| | - Yibai Xiong
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
| | - Zhiqi Song
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
| | - Yanhong Li
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
| | - Ling Zhang
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
| | - Chuan Qin
- Institute of Laboratory Animal Sciences, CAMS and Comparative Medicine Center, PUMC, Beijing, China
- Changping National Laboratory (CPNL), Beijing, China
| |
Collapse
|
22
|
Al-Sammarraie MR, Azaiez FD, Litaiem H. mRNA Vaccination for COVID-19 Lowers the Risk for Pulmonary Fibrosis Through GIP-10/Gal-3/HIF-1 Downregulation. Cureus 2025; 17:e84269. [PMID: 40525031 PMCID: PMC12168844 DOI: 10.7759/cureus.84269] [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] [Accepted: 05/17/2025] [Indexed: 06/19/2025] Open
Abstract
Background Pulmonary fibrosis (PF) is associated with coronavirus disease 2019 (COVID-19) through the occurrence of acute respiratory distress syndrome (ARDS). The overall sequence results in the elevation of the inflammation profile of infected individuals. The risk of PF onset in COVID-19 patients is not limited to the infection course but extends to post-infection periods. Gamma-inducible protein-10 (GIP-10) is a chemokine; its production and release are induced by interferon-gamma (IFN-γ). Objectives In this study, we aimed to investigate the role of GIP-10, Galectin-3 (Gal-3), and hypoxia inducible factor 1 (HIF-1) in PF-associated COVID-19 and the effectiveness of the Pfizer vaccine against the progression of PF and inflammation through evaluating these three biomarkers and their correlation with a few hematological parameters. Design & methods The study included 120 subjects (34-68 years) from Ibn Al-Nafees Hospital (Baghdad, Iraq). Three groups of 40 subjects were designed for our investigation as control, non-vaccinated COVID-19, and vaccinated COVID-19 patients. The presence of PF was evaluated in each participant. The COVID-19 patients with chronic kidney disease, liver cirrhosis, cancer, and pregnant women were excluded from this study. The GIP-10, Gal-3, and HIF-1 were evaluated in the subjects' serum using Sandwich ELISA technology. Results The results have shown significantly elevated levels of GIP-10, Gal-3, and HIF-1 in vaccinated and non-vaccinated PF-associated COVID-19 patients compared to the control, but the vaccinated patients exhibited significantly (p<0.05) lower levels of GIP-10, Gal-3, and HIF-1 compared to non-vaccinated patients. Moreover, in non-vaccinated PF-associated COVID-19 patients, GIP-10 did not correlate significantly with any parameter, while in vaccinated patients, it was correlated positively with age, WBC, RBC, and ESR. All of GIP-10, Gal-3, and HIF-1 expressed Odds ratios (OR) 1< as risks for PF in COVID-19 patients and can be used excellently to predict PF-associated COVID-19. Conclusions The Pfizer vaccine for COVID-19 has a positive role in managing GIP-10 and, therefore, better controls patients' inflammation profiles.
Collapse
Affiliation(s)
- Maha R Al-Sammarraie
- Department of Chemistry, University of Baghdad, Baghdad, IRQ
- Laboratory of Inorganic Chemistry, Faculty of Sciences, University of Sfax, Sfax, TUN
| | - Fatma D Azaiez
- Faculty of Pharmacy, University of Monastir, Monastir, TUN
| | - Hejer Litaiem
- Department of Chemistry, University of Sfax, Sfax, TUN
| |
Collapse
|
23
|
Shang Z, Huang L, Qin S. The underlying mechanism behind the different outcomes of COVID-19 in children and adults. Front Immunol 2025; 16:1440169. [PMID: 40370452 PMCID: PMC12075420 DOI: 10.3389/fimmu.2025.1440169] [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: 05/29/2024] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, has affected hundreds of millions of people globally, resulting in millions of deaths. During this pandemic, children have demonstrated greater resistance than adults, exhibiting lower infection rates, reduced mortality, and milder symptoms. Summarizing the differences in resistance between children and adults during COVID-19 can provide insights into protective mechanisms and potential implications for future treatments. In this review, we focused on summarizing and discussing the mechanisms for better protection of children in COVID-19. These protective mechanisms encompass several factors: the baseline expression of cell surface receptor ACE2 and hydrolase TMPRSS2, the impact of complications on COVID-19, and age-related cytokine profiles. Additionally, differences in local and systemic immune responses between children and adults also contribute significantly, particularly interferon responses, heterologous protection from non-COVID-19 vaccinations, and immune status variations influenced by micronutrient levels. The advantageous protection mechanisms of these children may provide insights into the prevention and treatment of COVID-19. Importantly, while age-related metabolic profiles and differential COVID-19 vaccine responses may contribute to protection in children, current comparative research remains limited and requires further investigation.
Collapse
Affiliation(s)
- Zifang Shang
- Research Experiment Center, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, Guangdong, China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou People's Hospital, Meizhou, Guangdong, China
| | - Ling Huang
- Department of Critical Medicine, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shijie Qin
- Innovative Vaccine and Immunotherapy Research Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Paediatric Research Institute, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
24
|
Parizad R, Batta A, Hatwal J, Taban-Sadeghi M, Mohan B. Emerging risk factors for heart failure in younger populations: A growing public health concern. World J Cardiol 2025; 17:104717. [PMID: 40308622 PMCID: PMC12038706 DOI: 10.4330/wjc.v17.i4.104717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
Heart failure (HF) is a growing public health concern, with an increasing incidence among younger populations. Traditionally, HF was considered a condition primarily affecting the elderly, but of late, emerging evidence hints at a rapidly rising HF incidence in youth in the past 2 decades. HF in youth has been linked to a complex interaction between emerging risk factors, such as metabolic syndrome, environmental exposures, genetic predispositions, and lifestyle behaviors. This review examines these evolving determinants, including substance abuse, autoimmune diseases, and the long-term cardiovascular effects of coronavirus disease 2019, which disproportionately affect younger individuals. Through a comprehensive analysis, the study highlights the importance of early detection, targeted prevention strategies, and multidisciplinary management approaches to address this alarming trend. Promoting awareness and integrating age-specific interventions could significantly reduce the burden of HF and improve long-term outcomes among younger populations.
Collapse
Affiliation(s)
- Razieh Parizad
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz 51656-87386, Iran
| | - Akash Batta
- Department of Cardiology, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India.
| | - Juniali Hatwal
- Department of Internal Medicine, Post Graduate Institute of Medical Education & Research, Chandigarh 160012, India
| | | | - Bishav Mohan
- Department of Cardiology, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India
| |
Collapse
|
25
|
Castro-Trujillo S, Castro-Meneses J, Rojas MC, Castro-Amaya M, Lastra G, Narváez CF. Regulatory cytokines modulate early isotype-specific response associated with COVID-19 survival. Front Immunol 2025; 16:1543626. [PMID: 40342417 PMCID: PMC12058664 DOI: 10.3389/fimmu.2025.1543626] [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: 02/07/2025] [Accepted: 04/01/2025] [Indexed: 05/11/2025] Open
Abstract
Identifying immune markers driving early and effective antibody response in patients with severe coronavirus disease 2019 (COVID-19) is critical due to the threat of future coronavirus pandemics, incomplete global vaccination, and suboptimal booster coverage. Patients with life-threatening severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are characterized by dysregulated thromboinflammation and cytokine storm that could influence the isotype virus-specific antibody response and the subsequent clinical outcome. We investigated the association between COVID-19-related mortality with the dynamics, magnitude, and relative avidity of nucleoprotein (N), spike (S), and receptor-binding domain (RBD)-specific IgM, IgA, and IgG in circulation. We also assessed the relationship between the virus-specific antibody responses and cytokine patterns, as well as systemic and pulmonary thromboinflammation markers. This multicenter study included COVID-19 patients hospitalized early in the pandemic, classified as survivors (n=62) and non-survivors (n=17). We developed indirect enzyme-linked immunosorbent assays (ELISAs) to evaluate each virus-specific isotype using well-characterized outpatient COVID-19 (n=180) and pre-pandemic cohorts (n=111). The pro-inflammatory interleukin (IL)-6 and tumor necrosis factor (TNF)-α, as well as the regulatory IL-10, transforming growth factor (TGF)-β1, and soluble tumor necrosis factor receptor I (sTNFRI) levels were evaluated. The ELISAs performed highly for all virus-specific isotypes, although modest for IgM-N. Non-survivors increased N-specific, but no S-specific, IgM and IgA responses throughout the disease course and, more notably, a delayed class switching to IgG-S and IgG-RBD compared to survivors. No differences were observed in the virus-specific IgG relative avidity. Survivors exhibited an antibody response proportional to the degree of systemic and pulmonary thromboinflammation, whereas non-survivors showed those dissociated because of their uncontrolled severe thromboinflammation. Only the survivors showed a dominant regulatory cytokine pattern in the early phase of infection (<10 days after symptoms onset), which strongly correlated with developing IgG-S and IgG-RBD protective antibodies. We developed easy-to-use immune assays that enable patient monitoring and identify at-risk populations in low- to middle-income regions. Non-survivors displayed an ineffective N-mediated antibody response, marked by an inability to control inflammation and a compromised time-dependent class switching toward S and RBD-specific IgG. The regulatory cytokine axis, including TGF-β1, maybe a critical immune correlate of effective antibody-mediated immunity in COVID-19.
Collapse
Affiliation(s)
- Sebastián Castro-Trujillo
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Juanita Castro-Meneses
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
- Programa de Biología Aplicada, Facultad de Ciencias Exactas y Naturales, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - María Clemencia Rojas
- Dirección Laboratorio de Salud Pública, Secretaría de Salud Departamental, Gobernación del Huila, Neiva, Huila, Colombia
| | - Marcela Castro-Amaya
- Departamento de Medicina Interna, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Giovani Lastra
- Departamento de Medicina Interna, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
- Servicio de Neumología, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Carlos F. Narváez
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| |
Collapse
|
26
|
Chatterjee D, Kurup D, Smeyne RJ. Environmental exposures and familial background alter the induction of neuropathology and inflammation after SARS-CoV-2 infection. NPJ Parkinsons Dis 2025; 11:86. [PMID: 40268936 PMCID: PMC12019605 DOI: 10.1038/s41531-025-00925-0] [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: 11/21/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Post-infection sequela of several viruses have been linked with Parkinson's disease (PD). Here, we investigated whether mice infected with SARS-CoV-2 alone or in combination with two putative Parkinsonian toxins, MPTP and paraquat, increased the susceptibility to develop Parkinsonian pathology. We also examined if G2019S LRRK2 mice had any change in sensitivity to SARS-CoV-2 as well as if vaccination against this virus altered any neuropathology. Infection with WA-1/2020 or Omicron B1.1.529 strains sensitized both WT and G2019S LRRK2 mice to the neuropathological effects of a subtoxic exposure to MPTP, but not paraquat. These neuropathologies were rescued in WT mice vaccinated with mRNA- or protein-based SARS-CoV-2 vaccines. However, G2019S LRRK2 mutant mice were only protected with the protein-based vaccine. These results highlight the role of both environmental exposures and familial background on the development of Parkinsonian pathology secondary to viral infection and the benefit of vaccines in reducing these risks.
Collapse
Affiliation(s)
- Debotri Chatterjee
- Department of Neurobiology, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA, 19107, USA
| | - Drishya Kurup
- Department of Microbiology and Immunology, Thomas Jefferson University, 233 S 10th Street, Philadelphia, PA, 19107, USA
- Jefferson Center for Vaccines and Pandemic Preparedness, 233 S 10th Street, Philadelphia, PA, 19107, USA
| | - Richard Jay Smeyne
- Department of Neurobiology, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA, 19107, USA.
| |
Collapse
|
27
|
Ghaffarpour S, Ghazanfari T, Ardestani SK, Naghizadeh MM, Vaez Mahdavi MR, Salehi M, Majd AMM, Rashidi A, Chenary MR, Mostafazadeh A, Rezaei A, Khodadadi A, Iranparast S, Khazaei HA. Cytokine profiles dynamics in COVID-19 patients: a longitudinal analysis of disease severity and outcomes. Sci Rep 2025; 15:14209. [PMID: 40269030 PMCID: PMC12019550 DOI: 10.1038/s41598-025-98505-y] [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: 11/06/2024] [Accepted: 04/11/2025] [Indexed: 04/25/2025] Open
Abstract
The outcome of the immune response depends on the content and magnitude of inflammatory mediators, the right time to start, and the duration of inflammatory responses. Patients with coronavirus disease 2019 (COVID-19) represent diverse disease severity. Understanding differences in immune responses in individuals with different disease severity levels can help elucidate disease mechanisms. Here, we serially analyzed the cytokine profiles of 809 patients with mild to critical COVID-19. The cytokine profile revealed an overall increase in IL-1β, IL-1Ra, TNF-α, IL-6, IL-2, IL-8, and IL-18 and impaired production of IFN-α and -β. Only an early rise in IL-1Ra, IL-6, and IL-2 levels was linked to worse disease outcomes. On the other hand, long-term rises in IL-1β, IL-1Ra, TNF-α, IL-6, IL-2, IL-8, and IL-18 levels were linked to worse disease outcomes. Principal component analysis identified a component, including IL-1β, TNF-α, IFN-α, and IL-12, that was associated with disease severity. Spearman analysis revealed that the correlation of IL-1β and IFN-α was entirely different between mild and critical patients. Therefore, the ratio of IL-1β to IFN-α seemed to be a suitable criterion for distinguishing critical patients from mild ones. The higher levels of the IL-1β to IFN-α ratio correlated with improved outcomes. These data point to an imbalance of IL-1β/IFNα, contributing to hyperinflammation in COVID-19.
Collapse
Affiliation(s)
- Sara Ghaffarpour
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran.
- Department of Immunology, Shahed University, Tehran, Iran.
| | - Sussan Kaboudanian Ardestani
- Immunoregulation Research Center, Shahed University, Tehran, Iran
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | | | - Mohammadreza Salehi
- Department of Infectious Diseases, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Azadeh Rashidi
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | | | - Amrollah Mostafazadeh
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Khodadadi
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sara Iranparast
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Ali Khazaei
- Department of Immunology and Internal Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
28
|
Palladini M, Mazza MG, Bravi B, Bessi M, Lorenzi MC, Spadini S, De Lorenzo R, Rovere-Querini P, Furlan R, Benedetti F. Sex-Specific Inflammatory Profiles Affect Neuropsychiatric Issues in COVID-19 Survivors. Biomolecules 2025; 15:600. [PMID: 40305313 PMCID: PMC12025053 DOI: 10.3390/biom15040600] [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: 03/10/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Post-COVID syndrome has unveiled intricate connections between inflammation, depressive psychopathology, and cognitive impairment. This study investigates these relationships in 101 COVID-19 survivors, focusing on sex-specific variations. Utilizing path modelling techniques, we analyzed the interplay of a one-month 48-biomarker inflammatory panel, with three-months of depressive symptoms and cognitive performance. The findings indicate that cognitive impairment is influenced by both inflammation and depression in the overall cohort. However, prominent sex-specific differences emerged. In females, a lingering imbalance between pro- and anti-inflammatory responses-likely reflecting the long-lasting immune alterations triggered by COVID-19-significantly affects cognitive functioning and shows a marginal, though not statistically significant, association with depressive symptoms. This suggests that a mixed inflammatory profile may contribute to these outcomes. Conversely, in males, inflammation was inversely associated with depression severity, with protective effects from regulatory mediators (IL-2, IL-4, IL-6, IL-15, LIF, TNF-α, β-NGF) against depression. In males, cognitive impairment appeared to be driven mainly by depressive symptoms, with minimal influence from inflammatory markers. These results highlight distinct sex-specific pathways in immune and inflammatory responses post-COVID-19, potentially shaped by endocrine mechanisms. The findings suggest that persistent inflammation may foster long-term neuropsychiatric sequelae, possibly through its effects on the brain, and underscore the need for sex-tailored therapeutic strategies to address the lasting impact of COVID-19.
Collapse
Affiliation(s)
- Mariagrazia Palladini
- Vita-Salute San Raffaele University, 20132 Milano, Italy; (R.D.L.); (P.R.-Q.); (R.F.); (F.B.)
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milano, Italy; (M.G.M.); (B.B.); (M.B.); (M.C.L.); (S.S.)
| | - Mario Gennaro Mazza
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milano, Italy; (M.G.M.); (B.B.); (M.B.); (M.C.L.); (S.S.)
| | - Beatrice Bravi
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milano, Italy; (M.G.M.); (B.B.); (M.B.); (M.C.L.); (S.S.)
| | - Margherita Bessi
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milano, Italy; (M.G.M.); (B.B.); (M.B.); (M.C.L.); (S.S.)
| | - Maria Cristina Lorenzi
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milano, Italy; (M.G.M.); (B.B.); (M.B.); (M.C.L.); (S.S.)
| | - Sara Spadini
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milano, Italy; (M.G.M.); (B.B.); (M.B.); (M.C.L.); (S.S.)
| | - Rebecca De Lorenzo
- Vita-Salute San Raffaele University, 20132 Milano, Italy; (R.D.L.); (P.R.-Q.); (R.F.); (F.B.)
- Unit of Innate Immunity and Tissue Remodelling, Department of Internal Medicine, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, 20132 Milano, Italy
| | - Patrizia Rovere-Querini
- Vita-Salute San Raffaele University, 20132 Milano, Italy; (R.D.L.); (P.R.-Q.); (R.F.); (F.B.)
- Unit of Innate Immunity and Tissue Remodelling, Department of Internal Medicine, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, 20132 Milano, Italy
| | - Roberto Furlan
- Vita-Salute San Raffaele University, 20132 Milano, Italy; (R.D.L.); (P.R.-Q.); (R.F.); (F.B.)
- Clinical Neuroimmunology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, 20132 Milano, Italy
| | - Francesco Benedetti
- Vita-Salute San Raffaele University, 20132 Milano, Italy; (R.D.L.); (P.R.-Q.); (R.F.); (F.B.)
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milano, Italy; (M.G.M.); (B.B.); (M.B.); (M.C.L.); (S.S.)
| |
Collapse
|
29
|
Wang J, Niu H, Kang J, Liu H, Dong X. Macrophage Polarization in Lung Diseases: From Mechanisms to Therapeutic Strategies. Immunol Invest 2025:1-27. [PMID: 40213814 DOI: 10.1080/08820139.2025.2490898] [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: 04/24/2025]
Abstract
Macrophages are pivotal immune cells involved in maintaining immune homeostasis and defending against pathogens. They exhibit significant plasticity and heterogeneity, enabling polarization into pro-inflammatory M1 or anti-inflammatory M2 phenotypes in response to distinct microenvironmental cues. The process of macrophage polarization is tightly regulated by complex signaling pathways and transcriptional networks. This review explores the factors influencing macrophage polarization, the associated signaling pathways, and their roles in the pathogenesis of lung diseases, including fibrosis, cancer, and chronic inflammatory conditions. By summarizing recent advances, we aim to provide insights into the immunoregulatory functions of macrophages and their therapeutic potential. Based on our review, it is believed that targeting macrophage polarization emerges as a promising approach for developing effective treatments for lung diseases, balancing inflammation and repair while mitigating disease progression.
Collapse
Affiliation(s)
- Jia Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Huajie Niu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Junwei Kang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Haiping Liu
- Department of Radiology, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, P.R. China
| | - Xiaoyang Dong
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
30
|
Levy E, Gilliaux G, Sarlet M, Desmecht D, Van Laere AS. Host-Strain-Specific Responses to Pneumonia Virus of Mice Infection: A Study of Lesions, Viral Load, and Cytokine Expression. Viruses 2025; 17:548. [PMID: 40284991 PMCID: PMC12031304 DOI: 10.3390/v17040548] [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: 02/24/2025] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Pneumonia virus of mice (PVM) infection is a reference animal model for human respiratory syncytial virus (hRSV), a leading cause of lower respiratory tract disease in children under 5 years of age and in the elderly. This longitudinal study employed necropsy to examine macroscopic lesions, histological slides to assess microscopic lesions, and qRT-PCR to measure lung viral load and cytokine expression in PVM-infected mice from three different genetic backgrounds, spanning from day 1 to day 6 post-infection. Our analysis reveals a strong correlation between viral load and microscopic lesions across the 129/Sv, BALB/c, and SJL/J mouse lines, indicating that PVM pathogenicity is partially driven by the virus itself. Additionally, a significant correlation between cytokine levels and lesion severity was observed in 129/Sv and BALB/c mice, suggesting an important role of cytokines in disease progression. This study emphasizes the interplay between viral load and cytokine-driven tissue damage, with genetic background significantly influencing disease outcomes.
Collapse
|
31
|
Dey P, Mal N, Sinha R, Kumar A, Kumar P, Saroj U, Chaudhuri PK, Sinha MBK, Guria R, Mishra B, Prasad ML, Kumar D, Kumar S, Prasad MK. Neutrophil gelatinase-associated lipocalin as a predictive biomarker of acute kidney injury in COVID-19 infection: A systematic review and meta-analysis. J Family Med Prim Care 2025; 14:1194-1206. [PMID: 40396107 PMCID: PMC12088576 DOI: 10.4103/jfmpc.jfmpc_1513_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 05/22/2025] Open
Abstract
Background Coronavirus 2019 (COVID-19) is an infectious disease caused by a novel coronavirus, SARS-CoV-2. Acute kidney injury (AKI) affects approximately 20-40% of patients with COVID-19 admitted to the intensive care unit (ICU), and it is a complication that has been linked to increased morbidity and mortality. Neutrophil gelatinase-associated lipocalin (NGAL) is a biomarker of acute kidney injury. Methods Articles were searched from databases such as PubMed, Google Scholar, and Cochrane Library till June 2023. Pooled sensitivity, specificity, area under the curve (AUC), diagnostic odds ratio (DOR), and summery receiver operating curve (SROC) were calculated with 95% confidence interval. I2 statistics and Chi-square test were used to look for the heterogeneity in between studies. Meta-regression was conducted to look for the source of heterogeneity and GRADE analysis was performed to look for the certainty of evidence. Results Altogether, eight studies were selected (4 serum/5 urine), out of which one study had both serum and urine NGAL data. The total sample size was 1,067 (349 serum/718 urine). For serum and urine NGAL, the pooled sensitivity was 0.79 (95% CI: 0.72-0.84) and 0.75 (95% CI: 0.68-0.80), pooled specificity was 0.87 (95% CI: 0.81-0.91) and 0.85 (95% CI: 0.77-0.91), DOR was 24 (95% CI: 14-43), and 17 (95% CI: 9-32) and AUC was 0.90 (95% CI: 0.87-0.92) and 0.80 (95% CI: 0.76-0.83), respectively. Conclusion Both serum and urine NGAL have favourable pooled sensitivity, specificity, DOR and AUC for the diagnosis of AKI in COVID-19 infection, however, with low certainty of evidence.
Collapse
Affiliation(s)
- Puja Dey
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Nilanjan Mal
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Rashmi Sinha
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Amit Kumar
- Department of Laboratory Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Pramod Kumar
- Department of Biochemistry, Hi-Tech Medical College and Hospital, Rourkela, Odisha, India
| | - Usha Saroj
- Department of Blood Centre and Transfusion Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Partha Kumar Chaudhuri
- Department of Paediatrics, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | | | - Rishi Guria
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Brajesh Mishra
- Department of TB and Chest, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Manohar Lal Prasad
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Divakar Kumar
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Satish Kumar
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Manoj Kumar Prasad
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| |
Collapse
|
32
|
Holland J, Sheehan D, Brown S, O'Flanagan S, Savinelli S, O'Keeffe F, Bramham J. Immune Response and Cognitive Impairment in Post-COVID Syndrome: A Systematic Review. Am J Med 2025; 138:698-711.e2. [PMID: 39362575 DOI: 10.1016/j.amjmed.2024.09.022] [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: 08/25/2023] [Revised: 07/23/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Altered immune response and cognitive difficulties have been demonstrated in studies of post-COVID syndrome, including differences in immune status and cognitive functioning in the months following infection. This review aimed to examine immune status and cognitive differences in post-COVID syndrome 12 or more weeks after COVID-19 infection. A further aim of this review was to explore a link between immune response and the cognitive deficits observed in this group. METHODS A systematic review was carried out using PubMed, PsychInfo, EMBASE, and Web of Science electronic databases of observational studies 12+ weeks after COVID-19 infection, with assessment of immune status and cognitive function in post-COVID syndrome samples. This review protocol was recorded on PROSPERO with registration number CRD42022366920. RESULTS Following eligibility screening, 11 studies met inclusion criteria and were selected for our review. Six of eight studies that examined between-group differences in specific domains suggested impaired cognition in the post-COVID syndrome population, with the domain of executive function particularly affected. Of 11 studies with immune data, 7 studies reported increased markers of inflammation in the post-COVID syndrome group, when compared with an age- and sex-matched "healthy control" sample, or population norms. Finally, when immune function and cognition are examined together, 6 studies presented results indicating a significant association between elevated immune response and cognitive function in post-COVID syndrome. CONCLUSION This review highlights the frequency of cognitive difficulties months after COVID-19 infection and explores heightened immune response as a predictor of this change. Six studies suggest that immune status is a predictor of cognitive function, examining a marker of immune function and objective cognitive performance at 12 or more weeks following infection. Future studies of cognitive function in post-COVID syndrome are needed to explore this relationship and underlying mechanisms leading to changes in cognitive performance.
Collapse
Affiliation(s)
| | | | | | - Susan O'Flanagan
- Department of Psychology, St Vincent's University Hospital, Dublin, Ireland; University College Dublin, Ireland
| | - Stefano Savinelli
- University College Dublin, Ireland; Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Fiadhnait O'Keeffe
- Department of Psychology, St Vincent's University Hospital, Dublin, Ireland; University College Dublin, Ireland
| | | |
Collapse
|
33
|
Tomos I, Grigoropoulos I, Kosti C, Chrysikos S, Digalaki A, Thomas K, Hillas G, Kazakou P, Antoniadou A, Kavatha D, Dimakou K. Comparison of effectiveness and safety between baricitinib and tocilizumab in severe COVID-19: a retrospective study. Expert Rev Respir Med 2025; 19:389-397. [PMID: 40017107 DOI: 10.1080/17476348.2025.2473486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Immunomodulators tocilizumab and baricitinib have been used for the treatment of severe COVID-19, however, there are only few published studies comparing their efficacy. RESEARCH DESIGN AND METHODS All consecutive non-ICU hospitalized severe COVID-19 patients who received baricitinib or tocilizumab, were included retrospectively. Primary outcomes were mortality or intubation on day 14, time to oxygen therapy weaning and duration of hospitalization. Safety was measured as treatment-related adverse events. RESULTS 321 hospitalized patients with severe COVID-19 were included (mean age 62.4 years ± 14.7); 241 (75.1%) received baricitinib (mean age 64.2 years ± 15.2) and 80 (24.9%) tocilizumab (mean age 57.3 ± 11.7). Patients who received baricitinib presented significantly lower risk of mortality or intubation on day 14, compared to the tocilizumab group after adjusting for age, sex, vaccination, Charlson comorbidity index, body mass index, remdesivir administration and WHO ordinal scale at enrollment (OR: 0.42, 95% CI: 0.20-0.86). In the augmented inverse-probability weighting regression, the protective role of baricitinib remained statistically significant (OR: 0.76, 95% CI: 0.66-0.88). No difference in secondary bacterial infections was detected, but tocilizumab was associated with significant higher rate of liver injury (Odds Ratio, 95%CI, p < 0.001). CONCLUSIONS Our study suggests survival and safety are significantly better for baricitinib compared to tocilizumab in severe COVID-19. Clinical randomized trials are needed for confirmation.
Collapse
Affiliation(s)
- Ioannis Tomos
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Ioannis Grigoropoulos
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Chrysavgi Kosti
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Serafeim Chrysikos
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Antonia Digalaki
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Konstantinos Thomas
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Georgios Hillas
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Pinelopi Kazakou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Anastasia Antoniadou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Dimitra Kavatha
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Katerina Dimakou
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| |
Collapse
|
34
|
Zhou F, Guo Y, Li W, Hu Y, Yang L, Fu S, Bao X, Tong H, Ye Y, Ding Z. Tetrastigma hemsleyanum polysaccharide protects against "two-hit" induced severe pneumonia via TLR4/NF-κB signaling pathway. Int J Biol Macromol 2025; 303:140639. [PMID: 39909274 DOI: 10.1016/j.ijbiomac.2025.140639] [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: 11/10/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Severe pneumonia, frequently accompanied by cytokine storms, stands as a perilous respiratory condition with alarmingly high mortality rates. Tetrastigma hemsleyanum polysaccharide (THP), a pivotal constituent derived from Tetrastigma hemsleyanum Diels et Gilg (TH), has demonstrated efficacy in treating lung inflammation. However, its precise efficacy and underlying mechanisms in the context of severe pneumonia remain elusive. Our research aims to elucidate THP's protective effects in a "two-hit" severe pneumonia model. Our observations indicate that THP administration markedly shields the lungs from injury, reduces pulmonary apoptosis, balances the formation of immune thrombus and alleviates oxidative stress in pneumonia-induced mice. Furthermore, THP significantly decreases the levels of pro-inflammatory cytokines, suggesting its robust anti-inflammatory capabilities. Notably, THP also plays a crucial role in normalizing gut microbiota imbalance, which is vital in the pathogenesis of severe pneumonia. Metabolomic analysis further validates THP's restorative effects on plasma metabolites, indicating its involvement in regulating energy metabolism and immune homeostasis. Mechanistically, THP targets the TLR4/NF-κB signaling pathway, a core mediator of inflammation, thereby dampening the inflammatory cascade. In summary, our findings underscore that THP, through its multifaceted actions targeting inflammation, oxidative stress, immune thrombus formation, gut microbiota regulation, and metabolic modulation, emerges as a promising therapeutic approach for severe pneumonia. This study provides invaluable insights into the potential applications of natural polysaccharides in treating severe pneumonia and highlights the significance of the TLR4/NF-κB pathway in the disease's progression.
Collapse
Affiliation(s)
- Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ying Guo
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wenxuan Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yiwen Hu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Liu Yang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Siyu Fu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Xiaodan Bao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongbin Tong
- Hangzhou HealthBank Medical Laboratory Co., Ltd., Hangzhou, Zhejiang 310053, China
| | - Yujian Ye
- Department of Dermatology, Third People's Hospital of Hangzhou, Hangzhou, China.
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
35
|
Ciurariu E, Balteanu MA, Georgescu M, Drăghici GA, Vlăsceanu SG, Șerb AF, Cioboată R. Left vs. Right Bundle Branch Block in COVID-19 Patients: Distinct Clinical Presentations and Prognostic Implications. J Clin Med 2025; 14:2310. [PMID: 40217761 PMCID: PMC11989766 DOI: 10.3390/jcm14072310] [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: 03/03/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: COVID-19 is associated with multiple systemic effects, including cardiovascular complications. However, its interplay with cardiac conduction abnormalities remains underexplored. We compared the clinical profile of COVID-19 patients with pre-existing left bundle branch block (LBBB) or right bundle branch block (RBBB) at hospital admission. Methods: This study included 100 COVID-19 patients with antecedent BBB (50 LBBB, 50 RBBB). Critical cardiometabolic, renal, hematological, and inflammatory markers were measured. Logistic regression was used to identify key predictors differentiating COVID-19 patients with LBBB and RBBB. Spearman's correlations were applied to assess intra-strata associations for these variables. Results: COVID-19 patients with LBBB patients were significantly more likely to display lower systolic blood pressure (p = 0.012) but greater left atrial size (p = 0.008), left ventricular diameter (p = 0.001), and interventricular septal thickness (p = 0.023). Hematological and inflammatory markers differed, with LBBB patients being prone to exhibit higher red cell distribution width (p = 0.005), lymphocyte count (p < 0.001), neutrophil count (p = 0.045), and C-reactive protein (p < 0.001). This group also tended to show lower erythrocyte sedimentation rate (p = 0.013) and glycated hemoglobin (p = 0.045) but higher random glucose (p = 0.014). Absolute lymphocyte count, C-reactive protein, and left ventricular diameter were the most robust predictors distinguishing LBBB from RBBB. Significant associations were found exclusively for LBBB, all of them being weak. These predominantly negative relationships indicated an inflammatory origin, and most of them occurred for lymphocyte count. Conclusions: COVID-19 patients with LBBB and RBBB present distinct clinical profiles at hospital admission. The former group demonstrates a more adverse baseline clinical profile, particularly in terms of cardiac and inflammatory markers. These findings suggest that pre-existing BBB type may influence disease progression, potentially helping in risk stratification for COVID-19 patients.
Collapse
Affiliation(s)
- Elena Ciurariu
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania; (E.C.); (M.G.)
| | - Mara Amalia Balteanu
- Department of Pulmonology, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
| | - Marius Georgescu
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania; (E.C.); (M.G.)
- Center of Immuno-Physiology and Biotechnologies (CIFBIOTEH), “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
| | - George Andrei Drăghici
- Department of Toxicology, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Silviu Gabriel Vlăsceanu
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alina-Florina Șerb
- Department of Biochemistry and Pharmacology, Biochemistry Discipline, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
| | - Ramona Cioboată
- Pneumology Department, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| |
Collapse
|
36
|
Plazas E, Sierra-Marquez L, Olivero-Verbel J. Bioactive Molecules from Tropical American Plants: Potential Anti-Inflammatory Agents for Cytokine Storm Management. Molecules 2025; 30:1486. [PMID: 40286093 PMCID: PMC11990702 DOI: 10.3390/molecules30071486] [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: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/29/2025] Open
Abstract
The cytokine storm, a hyperinflammatory response characterized by the excessive release of pro-inflammatory mediators such as TNFα, INFγ, IL-1β, IL-6, and GM-CSF, has been identified as a critical factor in the progression and severity of acute inflammatory conditions. Regulating these pathways is essential for mitigating systemic damage and improving outcomes. Natural products from tropical American plants have shown significant potential in modulating these hyperinflammatory responses. Key polyphenols, like quercetin and luteolin, found in plants such as Achyrocline satureioides and Mangifera indica demonstrate the downregulation of NF-κB and inhibition of pro-inflammatory cytokines. Alkaloids, such as berberine and mitraphylline, isolated from Berberis species and Uncaria tomentosa, respectively, have shown potent effects in suppressing nitric oxide production and regulating inflammasomes. Terpenoids, including parthenolide from Tanacetum parthenium and curcumol from Curcuma longa, exhibit multitarget activity, reducing cytokine levels and inhibiting key inflammatory enzymes like COX-2 and iNOS. These findings highlight the immense potential of bioactive compounds from tropical American plants as modulators of immune-inflammatory pathways, providing a foundation for developing effective therapeutic agents to counteract the severe effects of cytokine storms.
Collapse
Affiliation(s)
| | | | - Jesus Olivero-Verbel
- Environmental and Computational Chemistry Group, School of Pharmaceutical Sciences, University of Cartagena, Cartagena 130014, Colombia; (E.P.); (L.S.-M.)
| |
Collapse
|
37
|
Li Y, Jiao J, Qiao H, Wang C, Li L, Jin F, Ye D, Chen Y, Zhang Q, Li M, Zhao Z, Zhang J, Wang L. Aromatic Molecular Compatibility Attenuates Influenza Virus-Induced Acute Lung Injury via the Lung-Gut Axis and Lipid Droplet Modulation. Pharmaceuticals (Basel) 2025; 18:468. [PMID: 40283905 PMCID: PMC12030469 DOI: 10.3390/ph18040468] [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: 03/08/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Acute lung injury (ALI) is a major cause of death in patients with various viral pneumonias. Our team previously identified four volatile compounds from aromatic Chinese medicines. Based on molecular compatibility theory, we defined their combination as aromatic molecular compatibility (AC), though its therapeutic effects and underlying mechanisms remain unclear. Methods: This study used influenza A virus (IAV) A/PR/8/34 to construct cell and mouse models of ALI to explore AC's protective effects against viral infection. The therapeutic effect of AC was verified by evaluating the antiviral efficacy in the mouse models, including improvements in their lung and colon inflammation, oxidative stress, and the suppression of the NLRP3 inflammasome. In addition, 16S rDNA and lipid metabolomics were used to analyze the potential therapeutic mechanisms of AC. Results: Our in vitro and in vivo studies demonstrated that AC increased the survival of the IAV-infected cells and mice, inhibited influenza virus replication and the expression of proinflammatory factors in the lung tissues, and ameliorated barrier damage in the colonic tissues. In addition, AC inhibited the expression of ROS and the NLRP3 inflammasome and improved the inflammatory cell infiltration into the lung tissues. Finally, AC effectively regulated intestinal flora disorders and lipid metabolism in the model mice, significantly reduced cholesterol and triglyceride expression, and thus reduced the abnormal accumulation of lipid droplets (LDs) after IAV infection. Conclusions: In this study, we demonstrated that AC could treat IAV-induced ALIs through multiple pathways, including antiviral and anti-inflammatory pathways and modulation of the intestinal flora and the accumulation of LDs.
Collapse
Affiliation(s)
- Yi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| | - Jiakang Jiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Haoyi Qiao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| | - Conghui Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| | - Linze Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| | - Fengyu Jin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| | - Danni Ye
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| | - Yawen Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| | - Qi Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Zhongpeng Zhao
- Beijing Minhai Biotechnology Co., Ltd., Beijing 102600, China
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Linyuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.L.)
| |
Collapse
|
38
|
He MZ, Zhang HT, Yang Y, Fang Y, Zhang M, Deng SQ, Sun X. Coinfection of COVID-19 and malaria: clinical profiles, interactions, and strategies for effective control. Malar J 2025; 24:99. [PMID: 40133914 PMCID: PMC11938571 DOI: 10.1186/s12936-025-05315-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Since SARS-CoV-2 has caused unprecedented changes in the epidemiology of other infectious diseases, investigations on coinfection between SARS-CoV-2 and one of the famous vector-borne diseases, malaria, are crucial for disease control, especially in malaria-endemic areas. The clinical profiles, possible mechanisms for interactions, and representative control measures of COVID-19 and malaria coinfections have recently garnered public attention. The overlap in epidemiology, infection incubation, and clinical symptoms between COVID-19 and malaria coinfections has been thoroughly discussed to provide a detailed diagnostic procedure for coinfections, thereby guiding appropriate clinical interventions. Immunological and genetic evidence has shown that previous malaria exposure may protect the body from the poor prognosis of COVID-19. ACE2 downregulation and TLR-induced pathways play a role in this protective effect, as do CD8 + and CD4 + T-cell activation and coinhibitory receptor upregulation, which help maintain a balance of immune reactions. Finally, multiple control measures for coinfections were discussed, and malaria control efforts were enriched in the context of COVID-19. These efforts included (1) developing vaccinations; (2) evaluating the efficacy of anti-malarial drugs in the SARS-CoV-2 treatment; (3) exploring recent advances in natural products that are potentially useful for coinfection treatment; (4) researching and implementing bioinsecticides for malaria control, such as gene-driven mosquitoes, fungi, and bacterial symbionts; and (5) improving national electronic disease surveillance platforms in malaria-endemic regions. At last, the above findings summarized valuable lessons about malaria and COVID-19 control and expedite further investigations on coinfections with complex clinical presentations.
Collapse
Affiliation(s)
- Mu-Zi He
- Gezhouba Central Hospital of Sinopharm, The Third Clinical Medical College of the Three Gorges University, Yichang, 443002, Hubei, China
- Department of Pathogen Biology, Anhui Province Key Laboratory of Zoonoses, The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Hai-Ting Zhang
- Department of Pathogen Biology, Anhui Province Key Laboratory of Zoonoses, The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yi Yang
- Gezhouba Central Hospital of Sinopharm, The Third Clinical Medical College of the Three Gorges University, Yichang, 443002, Hubei, China
| | - Yi Fang
- Gezhouba Central Hospital of Sinopharm, The Third Clinical Medical College of the Three Gorges University, Yichang, 443002, Hubei, China
| | - Mao Zhang
- Gezhouba Central Hospital of Sinopharm, The Third Clinical Medical College of the Three Gorges University, Yichang, 443002, Hubei, China
| | - Sheng-Qun Deng
- Department of Pathogen Biology, Anhui Province Key Laboratory of Zoonoses, The Provincial Key Laboratory of Zoonoses of High Institutions in Anhui, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
- Department of Pathology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| | - Xun Sun
- Gezhouba Central Hospital of Sinopharm, The Third Clinical Medical College of the Three Gorges University, Yichang, 443002, Hubei, China.
| |
Collapse
|
39
|
Brügger M, Machahua C, Zumkehr T, Cismaru C, Jandrasits D, Trüeb B, Ezzat S, Oliveira Esteves BI, Dorn P, Marti TM, Zimmer G, Thiel V, Funke-Chambour M, Alves MP. Aging shapes infection profiles of influenza A virus and SARS-CoV-2 in human precision-cut lung slices. Respir Res 2025; 26:112. [PMID: 40128814 PMCID: PMC11934781 DOI: 10.1186/s12931-025-03190-0] [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: 10/23/2024] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) outbreak revealed the susceptibility of elderly patients to respiratory virus infections, showing cell senescence or subclinical persistent inflammatory profiles and favoring the development of severe pneumonia. METHODS In our study, we evaluated the potential influence of lung aging on the efficiency of replication of influenza A virus (IAV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as well as determining the pro-inflammatory and antiviral responses of the distal lung tissue. RESULTS Using precision-cut lung slices (PCLS) from donors of different ages, we found that pandemic H1N1 and avian H5N1 IAV replicated in the lung parenchyma with high efficacy. In contrast to these IAV strains, SARS-CoV-2 Early isolate and Delta variant of concern (VOC) replicated less efficiently in PCLS. Interestingly, both viruses showed reduced replication in PCLS from older compared to younger donors, suggesting that aged lung tissue represents a suboptimal environment for viral replication. Regardless of the age-dependent viral loads, PCLS responded to H5N1 IAV infection by an induction of IL-6 and IP10/CXCL10, both at the mRNA and protein levels, and to H1N1 IAV infection by induction of IP10/CXCL10 mRNA. Finally, while SARS-CoV-2 and H1N1 IAV infection were not causing detectable cell death, H5N1 IAV infection led to more cytotoxicity and induced significant early interferon responses. CONCLUSIONS In summary, our findings suggest that aged lung tissue might not favor viral dissemination, pointing to a determinant role of dysregulated immune mechanisms in the development of severe disease.
Collapse
Affiliation(s)
- Melanie Brügger
- Institute of Virology and Immunology, Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| | - Carlos Machahua
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Trix Zumkehr
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Christiana Cismaru
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Institute of Virology, Freie Universitaet Berlin, Berlin, Germany
| | - Damian Jandrasits
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Spiez Laboratory, Federal Office for Civil Protection, Spiez, Switzerland
| | - Bettina Trüeb
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sara Ezzat
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Blandina I Oliveira Esteves
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas M Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Gert Zimmer
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Volker Thiel
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, Bern, Switzerland
- European Virus Bioinformatics Center, Jena, Germany
| | - Manuela Funke-Chambour
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marco P Alves
- Institute of Virology and Immunology, Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, Bern, Switzerland.
| |
Collapse
|
40
|
Szachowicz PJ, Wohlford-Lenane C, Donelson CJ, Ghimire S, Thurman A, Xue B, Boly TJ, Verma A, MašinoviĆ L, Bermick JR, Rehman T, Perlman S, Meyerholz DK, Pezzulo AA, Zhang Y, Smith RJ, McCray PB. Complement is primarily activated in the lung in a mouse model of severe COVID-19. iScience 2025; 28:111930. [PMID: 40034849 PMCID: PMC11875145 DOI: 10.1016/j.isci.2025.111930] [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: 07/15/2024] [Revised: 10/21/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
In vitro studies and observational human disease data suggest the complement system contributes to SARS-CoV-2 pathogenesis, although how complement dysregulation develops in severe COVID-19 is unknown. Here, using a mouse-adapted SARS-CoV-2 virus (SARS2-N501YMA30) and a mouse model of COVID-19, we identify significant serologic and pulmonary complement activation post-infection. We observed C3 activation in airway and alveolar epithelia, and pulmonary vascular endothelia. Our evidence suggests the alternative pathway is the primary route of complement activation, however, components of both the alternative and classical pathways are produced locally by respiratory epithelial cells following infection, and increased in primary cultures of human airway epithelia following cytokine and SARS-CoV-2 exposure. This tissue-specific complement response appears to precede lung injury and inflammation. Our results suggest that complement activation is a defining feature of severe COVID-19 in mice, agreeing with previous publications, and provide the basis for further investigation into the role of complement in COVID-19.
Collapse
Affiliation(s)
- Peter J. Szachowicz
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA 52242, USA
| | | | - Cobey J. Donelson
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Shreya Ghimire
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA 52242, USA
| | - Andrew Thurman
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA 52242, USA
| | - Biyun Xue
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
| | - Timothy J. Boly
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
| | - Abhishek Verma
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA 52242, USA
| | - Leila MašinoviĆ
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA 52242, USA
| | - Jennifer R. Bermick
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
| | - Tayyab Rehman
- Department of Inernal Medicine, University of Michigan, Division of Pulmonary and Critical Care Medicine, Ann Arbor, MI 48109, USA
| | - Stanley Perlman
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA 52242, USA
| | - David K. Meyerholz
- Department of Pathology, The University of Iowa, Iowa City, IA 52242, USA
| | - Alejandro A. Pezzulo
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA 52242, USA
| | - Yuzhou Zhang
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Richard J.H. Smith
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Paul B. McCray
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
41
|
Chuang CC, Liu YC, Ou YY. DeepEpiIL13: Deep Learning for Rapid and Accurate Prediction of IL-13-Inducing Epitopes Using Pretrained Language Models and Multiwindow Convolutional Neural Networks. ACS OMEGA 2025; 10:9675-9683. [PMID: 40092768 PMCID: PMC11904640 DOI: 10.1021/acsomega.4c10960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025]
Abstract
Accurate prediction of interleukin-13 (IL-13)-inducing epitopes is crucial for advancing targeted therapies against allergic inflammation, the cytokine storm associated with severe COVID-19, and related disorders. Current epitope prediction methods, however, often exhibit limitations in efficiency and accuracy. To address this, we introduce DeepEpilL13, a novel deep learning framework that uniquely synergizes pretrained language models with multiwindow convolutional neural networks (CNNs) for the rapid and accurate identification of IL-13-inducing epitopes from protein sequences. DeepEpilL13 leverages high-dimensional embeddings generated by the pretrained language model, which capture rich contextual information from protein sequences. These embeddings are then processed by a multiwindow CNN architecture, enabling the effective exploration of both local and global sequence patterns pertinent to IL-13 induction. The proposed DeepEpilL13 approach underwent rigorous evaluation using both benchmark data sets and an independent SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) data set. Results demonstrate that DeepEpilL13 achieves superior performance compared with traditional methods. On the benchmark data set, DeepEpilL13 attained a Matthews correlation coefficient (MCC) of 0.52 and an area under the receiver operating characteristic curve (AUC) of 0.86. Notably, when assessed on the independent SARS-CoV-2 data set, DeepEpilL13 exhibited remarkable robustness, achieving an MCC of 0.63 and an AUC of 0.92. These metrics underscore the enhanced predictive capability and robust applicability of DeepEpilL13, particularly within the context of the COVID-19 research and related viral infections. This study presents DeepEpilL13 as a powerful and efficient deep learning framework for accurate epitope prediction. By offering significant improvement in performance and robustness, DeepEpilL13 provides new and promising avenues for the development of epitope-based vaccines and immunotherapies specifically targeting IL-13-mediated disorders. The successful and rapid identification of IL-13-inducing epitopes using DeepEpilL13 paves the way for novel therapeutic interventions against a range of conditions, including allergic diseases, inflammatory conditions, and severe viral infections such as COVID-19, with potential for a significant impact on public health outcomes.
Collapse
Affiliation(s)
- Cheng-Che Chuang
- Department
of Computer Science and Engineering, Yuan
Ze University, Chung-Li 32003, Taiwan
| | - Yu-Chen Liu
- Department
of Computer Science and Engineering, Yuan
Ze University, Chung-Li 32003, Taiwan
| | - Yu-Yen Ou
- Department
of Computer Science and Engineering, Yuan
Ze University, Chung-Li 32003, Taiwan
- Graduate
Program in Biomedical Informatics, Yuan
Ze University, Chung-Li 32003, Taiwan
| |
Collapse
|
42
|
Alloza-Moral I, Aldekoa-Etxabe A, Tulloch-Navarro R, Fiat-Arriola A, Mar C, Urrechaga E, Ponga C, Artiga-Folch I, Garcia-Bediaga N, Aspichueta P, Martin C, Zarandona-Garai A, Pérez-Fernández S, Arana-Arri E, Triviño JC, Uranga A, España PP, Vandenbroeck-van-Caeckenbergh K. Genetic Analysis and Predictive Modeling of COVID-19 Severity in a Hospital-Based Patient Cohort. Biomolecules 2025; 15:393. [PMID: 40149929 PMCID: PMC11940120 DOI: 10.3390/biom15030393] [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: 12/18/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
The COVID-19 pandemic has had a devastating impact, with more than 7 million deaths worldwide. Advanced age and comorbidities partially explain severe cases of the disease, but genetic factors also play a significant role. Genome-wide association studies (GWASs) have been instrumental in identifying loci associated with SARS-CoV-2 infection. Here, we report the results from a >820 K variant GWAS in a COVID-19 patient cohort from the hospitals associated with IIS Biobizkaia. We compared intensive care unit (ICU)-hospitalized patients with non-ICU-hospitalized patients. The GWAS was complemented with an integrated phenotype and genetic modeling analysis using HLA genotypes, a previously identified COVID-19 polygenic risk score (PRS) and clinical data. We identified four variants associated with COVID-19 severity with genome-wide significance (rs58027632 in KIF19; rs736962 in HTRA1; rs77927946 in DMBT1; and rs115020813 in LINC01283). In addition, we designed a multivariate predictive model including HLA, PRS and clinical data which displayed an area under the curve (AUC) value of 0.79. Our results combining human genetic information with clinical data may help to improve risk assessment for the development of a severe outcome of COVID-19.
Collapse
Affiliation(s)
- Iraide Alloza-Moral
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Physiology Department, Faculty of Medicine and Nursery, Basque Country University (UPV/EHU), 48940 Leioa, Spain;
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
| | - Ane Aldekoa-Etxabe
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
| | - Raquel Tulloch-Navarro
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
| | - Ainhoa Fiat-Arriola
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
| | - Carmen Mar
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Eloisa Urrechaga
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Cristina Ponga
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Isabel Artiga-Folch
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
| | - Naiara Garcia-Bediaga
- Bioinformatic Unit, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (N.G.-B.); (A.Z.-G.); (S.P.-F.)
| | - Patricia Aspichueta
- Physiology Department, Faculty of Medicine and Nursery, Basque Country University (UPV/EHU), 48940 Leioa, Spain;
- Research Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), 28029 Madrid, Spain
- Biobizkaia Health Research Institute, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Cesar Martin
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Biochemistry and Molecular Biology Department, Science and Technology School, Basque Country University (UPV/EHU), 48940 Leioa, Spain
- Biofisika Institute (UPV/EHU, CSIC), 48940 Leioa, Spain
| | - Aitor Zarandona-Garai
- Bioinformatic Unit, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (N.G.-B.); (A.Z.-G.); (S.P.-F.)
| | - Silvia Pérez-Fernández
- Bioinformatic Unit, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (N.G.-B.); (A.Z.-G.); (S.P.-F.)
| | - Eunate Arana-Arri
- Clinical Epidemiology Unit, Biobizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces s/n, 48903 Barakaldo, Spain;
| | | | - Ane Uranga
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Pedro-Pablo España
- Pneumology Department, Galdakao-Usansolo University Hospital, Biobizkaia Health Research Institute, 48960 Galdakao, Spain; (C.M.); (E.U.); (C.P.); (A.U.); (P.-P.E.)
| | - Koen Vandenbroeck-van-Caeckenbergh
- Inflammation & Biomarkers Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.A.-M.); (A.A.-E.); (R.T.-N.); (A.F.-A.); (C.M.)
- Red de Enfermedades Inflamatorias (REI), Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Carlos IIII Health Research Institute, 28029 Madrid, Spain
- Biochemistry and Molecular Biology Department, Science and Technology School, Basque Country University (UPV/EHU), 48940 Leioa, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
43
|
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] [Download PDF] [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
- SARS-CoV-2/physiology
- SARS-CoV-2/immunology
- Mice
- COVID-19/immunology
- COVID-19/virology
- Virus Replication
- Receptors, Leptin/genetics
- Receptors, Leptin/metabolism
- Obesity/immunology
- Obesity/complications
- Signal Transduction
- Humans
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/complications
- Leptin/metabolism
- Interferon Type I/immunology
- Interferon Type I/metabolism
- Lung/immunology
- Lung/virology
- Immunity, Cellular
- Mice, Inbred C57BL
- Immunity, Innate
- Male
- Disease Models, Animal
- Antibodies, Neutralizing/blood
- Interferons
- 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
|
44
|
Adilović M. COVID-19 related complications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:259-314. [PMID: 40246346 DOI: 10.1016/bs.pmbts.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic has significantly impacted global healthcare systems, revealed vulnerabilities and prompted a re-evaluation of medical practices. Acute complications from the virus, including cardiovascular and neurological issues, have underscored the necessity for timely medical interventions. Advances in diagnostic methods and personalized therapies have been pivotal in mitigating severe outcomes. Additionally, Long COVID has emerged as a complex challenge, affecting various body systems and leading to respiratory, cardiovascular, neurological, psychological, and musculoskeletal problems. This broad spectrum of complications highlights the importance of multidisciplinary management approaches that prioritize therapy, rehabilitation, and patient-centered care. Vulnerable populations such as paediatric patients, pregnant women, and immunocompromised individuals face unique risks and complications, necessitating continuous monitoring and tailored management strategies to reduce morbidity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička cesta, Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
45
|
Torres-Poveda K, Bahena-Román M, Contreras-Ochoa CO, Lagunas-Martínez A, Bermúdez-Morales VH, Pando-Robles V, Ortiz-Flores E, Cortés-Pedroza F, Santana-Román ME, Martínez-Campos C, Sánchez-Alemán M, Manzo-Merino J, Morales-Ortega A, Madrid-González DA, Cantú-Cuevas MA, Barón-Olivares H, Madrid-Marina V. High nasopharyngeal and serum IL-6 levels and the - 573G > C polymorphism (rs1800796) are linked with the risk of severe COVID-19 in a Mexican population: a case‒control study. BMC Infect Dis 2025; 25:315. [PMID: 40045221 PMCID: PMC11884130 DOI: 10.1186/s12879-025-10695-y] [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: 02/24/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND COVID-19 was the leading cause of death in Mexico between 2020 and 2021. SARS-CoV-2 infection varies widely among individuals and populations. Since variations in genes related to the immune response may play a role in the susceptibility to and outcome of COVID-19, the associations of gene polymorphisms (SNPs) of IL-6 (- 573G > C, rs1800796), TNF-α (- 308G > A, rs1800629), and IFN-γ (- 1615 C > T, rs2069705) with the expression levels of these proteins in the nasopharynx and serum were evaluated in a Mexican population with mild, severe, or critical COVID-19. METHODS A total of 560 COVID-19 patients (309 mild, 163 severe, and 88 critical cases) and 560 age- and sex-matched COVID-19-negative controls were recruited for this case‒control study. The selected SNPs were genotyped via allelic discrimination. Logistic regression analysis was conducted considering four models of inheritance, and ORs were determined for each genotypic variant, adjusting for associated comorbidities in the multivariate model. The nasopharyngeal mRNA expression levels of IL-6, IFN-γ and TNF-α were determined. The levels of IL-6, IFN-γ, IFN-α2, and TNF-α in the serum were quantified. Significant differences were assessed via the Wilcoxon Mann‒Whitney U test. RESULTS The C allele of the IL-6 - 573 SNP was associated with a greater risk of mild and severe COVID-19 (OR: 2.3, CI: 1.897-2.838, p = 0.0001; and OR: 1.5, CI: 1.167-1.949, p = 0.002, respectively), whereas the A allele of the TNF-α - 308 SNP and the T allele of the IFN-γ - 1615 SNP were shown protective roles against severe COVID-19 (OR: 0.3, CI: 0.189-0.537, p = 0.0001; and OR: 0.7, CI: 0.563-1.006, p = 0.05) and against critical COVID-19 (OR: 0.3, CI: 0.158-0.640, p = 0.001; and OR: 0.4, CI: 0.290-0.678, p = 0.0001), adjusting for diabetes and hypertension. Nasopharyngeal IL-6 expression levels were lower in mild COVID-19 patients (p = 0.001) than in critical patients (p = 0.005). Serum IL-6 levels were significantly elevated in the critical cases (p = 0.01). CONCLUSIONS Our results revealed that the IL-6 - 573 G > C SNP and increased IL-6 nasopharyngeal and serum levels are associated with the risk of severe COVID-19 in a Mexican population.
Collapse
Affiliation(s)
- Kirvis Torres-Poveda
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
- Secretaria de Ciencia, Humanidades, Tecnología e Innovación (SECIHTI)-Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Margarita Bahena-Román
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Carla O Contreras-Ochoa
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Alfredo Lagunas-Martínez
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | | | - Victoria Pando-Robles
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Esmeralda Ortiz-Flores
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Fabiola Cortés-Pedroza
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - María E Santana-Román
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Cecilia Martínez-Campos
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Miguel Sánchez-Alemán
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Joaquin Manzo-Merino
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ausencio Morales-Ortega
- Laboratorio Estatal de Salud Pública. Health Services of the State of Morelos, Jiutepec, Mexico
| | | | | | - Héctor Barón-Olivares
- Dirección General de Coordinación y Supervisión. Health Services of the State of Morelos, Cuernavaca, Mexico
| | - Vicente Madrid-Marina
- Center for Research on Infectious Diseases, Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico.
| |
Collapse
|
46
|
Berkebile G, Barbé F, Malaplate C, Le Collen L, Guéant JL, Klein M, Oussalah A. Population-level impact of COVID-19 on thyroid function tests: Results from a repeated cross-sectional study. J Infect Public Health 2025; 18:102681. [PMID: 39874641 DOI: 10.1016/j.jiph.2025.102681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/11/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
PURPOSE Reports have highlighted thyroid abnormalities, including subacute thyroiditis and thyrotoxicosis, in COVID-19 patients, with a potential link between thyroid dysfunction and disease severity. However, population-level studies on COVID-19's impact on thyroid hormone levels are limited. We aimed to assess the impact of the COVID-19 pandemic on thyroid function tests at the population level. METHODS We conducted a repeated cross-sectional study on consecutive patients who underwent thyroid function tests between March 1 and April 15, 2020, during the first wave of COVID-19 in northeastern France, and compared the results with those from the same period in 2018 and 2019. RESULTS The study analyzed 3968 tests, including 1534 in 2018, 1547 in 2019, and 887 in 2020. Patients tested in the first wave of COVID-19 had significantly lower TSH and FT3 levels and higher FT4 levels than those in reference periods. On ROC analysis, the optimal thresholds for FT3 and FT4 were ≤ 4.5 pmol/L and > 12 pmol/L, respectively. On multivariable analysis, FT3 ≤ 4.5 pmol/L and FT4 > 12 pmol/L were independently associated with the first wave of COVID-19. The proportion of subjects with concurrent changes in FT3 and FT4 levels was significantly higher in 2020 than in the reference periods, with an odds ratio of 3.62 (95 % CI, 2.77-4.73). A phenome-wide association study of 128 clinical and biological predictors identified an independent association between a low FT3/FT4 ratio and COVID-19, hypertension, or amiodarone therapy, suggesting the contribution of euthyroid sick syndrome to this presentation. This association remained significant after adjustment for potential confounders such as thyroid disease, steroids, and iodinated contrast injection. CONCLUSION These findings suggest that COVID-19 is associated with significant population-level variation in thyroid function tests, which may have implications for managing COVID-19 patients.
Collapse
Affiliation(s)
- Gabriel Berkebile
- Department of Endocrinology, Diabetology, and Nutrition (EDN), University Hospital of Nancy, Nancy F-54000, France
| | - Françoise Barbé
- Department of Genomic Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy F-54000, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy F-54000, France
| | - Catherine Malaplate
- Department of Genomic Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy F-54000, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy F-54000, France; Faculty of Medicine of Nancy, University of Lorraine, Nancy F-54000, France
| | - Lauriane Le Collen
- Department of Genomic Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy F-54000, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy F-54000, France; Faculty of Medicine of Nancy, University of Lorraine, Nancy F-54000, France
| | - Jean-Louis Guéant
- Department of Genomic Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy F-54000, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy F-54000, France; Faculty of Medicine of Nancy, University of Lorraine, Nancy F-54000, France; INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Nancy F-54000, France
| | - Marc Klein
- Department of Endocrinology, Diabetology, and Nutrition (EDN), University Hospital of Nancy, Nancy F-54000, France; Faculty of Medicine of Nancy, University of Lorraine, Nancy F-54000, France.
| | - Abderrahim Oussalah
- Department of Genomic Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy F-54000, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy F-54000, France; Faculty of Medicine of Nancy, University of Lorraine, Nancy F-54000, France; INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Nancy F-54000, France.
| |
Collapse
|
47
|
Lemos FFB, Lopes LW, Brito GC, Viana AIS, de Castro CT, Luz MS, Gonçalves AP, Dórea RSDM, da Silva FAF, de Brito BB, Santos MLC, Júnior GMS, de Lorenzo Barcia MTA, de Amorim Marques R, Botelho AB, Dantas ACS, Pinheiro FD, Teixeira AF, Souza CL, Oliveira MV, de Magalhães Queiroz DM, de Melo FF. Prognostic significance of cytokine dysregulation in critically ill COVID-19 patients. Cytokine 2025; 187:156867. [PMID: 39874939 DOI: 10.1016/j.cyto.2025.156867] [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: 07/01/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Understanding the immunopathogenesis of COVID-19 has yielded valuable insights into predicting adverse outcomes-particularly mortality. However, significant gaps persist in our comprehension of the complex interplay among the proposed pathophysiological mechanisms. Here, we aim to investigate the immunological factors associated with mortality in critically ill, unvaccinated COVID-19 patients admitted to the intensive care unit (ICU). METHODS We conducted a single-center, prospective study involving 56 unvaccinated COVID-19 patients admitted to the ICU. Plasma cytokine levels at admission were quantified using enzyme-linked immunosorbent assay (ELISA). Continuous variables were presented as median (IQR), and categorical variables as frequencies and percentages. Non-parametric tests assessed group differences. Logistic regression and receiver operating characteristic (ROC) curve analyses identified predictors of mortality, with bootstrapping (1000 re-samplings; 95 % BCa CI) applied for model validation. RESULTS Deceased patients exhibited significantly higher levels of interleukin (IL)-1β, IL-2, IL-6, transforming growth factor (TGF)-β, and interferon (IFN)-γ compared to survivors. Conversely, IL-10 and IL-27 were associated with favorable outcomes. Logistic regression modeling identified elevated IL-2 and IFN-γ levels as significant predictors of mortality. Notably, individual ROC curve analyses demonstrated that IL-1β and TGF-β had excellent discriminatory ability for mortality, while IFN-γ, IL-2, and IL-27 showed very good to excellent discriminatory capacity. CONCLUSION Our results indicate that distinct cytokine profiles differentiate survivors from non-survivors in critically ill, unvaccinated COVID-19 patients. These findings highlight the importance of cytokine dysregulation in severe COVID-19 cases and suggest potential targets for prognostic approaches. Further research is warranted to validate these results and translate them into effective clinical management strategies.
Collapse
Affiliation(s)
- Fabian Fellipe Bueno Lemos
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Luana Weber Lopes
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Gabriel Carvalho Brito
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Airton Idalecio Sousa Viana
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Marcel Silva Luz
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - André Pereira Gonçalves
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | | - Breno Bittencourt de Brito
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Maria Luísa Cordeiro Santos
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | | | | - André Bezerra Botelho
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Anna Carolina Saúde Dantas
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fillipe Dantas Pinheiro
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Adriano Fernandes Teixeira
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Cláudio Lima Souza
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Márcio Vasconcelos Oliveira
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Fabrício Freire de Melo
- Multidisciplinary Health Institute, Federal University of Bahia, Vitória da Conquista 45029-094, Bahia, Brazil.
| |
Collapse
|
48
|
Nawa H, Murakami M. Neurobiology of COVID-19-Associated Psychosis/Schizophrenia: Implication of Epidermal Growth Factor Receptor Signaling. Neuropsychopharmacol Rep 2025; 45:e12520. [PMID: 39754403 PMCID: PMC11702486 DOI: 10.1002/npr2.12520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/06/2025] Open
Abstract
COVID-19 exhibits not only respiratory symptoms but also neurological/psychiatric symptoms rarely including delirium/psychosis. Pathological studies on COVID-19 provide evidence that the cytokine storm, in particular (epidermal growth factor) EGF receptor (EGFR, ErbB1, Her1) activation, plays a central role in the progression of viral replication and lung fibrosis. Of note, SARS-CoV-2 virus (specifically, S1 spike domain) mimics EGF and directly transactivates EGFR, preceding the inflammatory process. In agreement, the anticancer drugs targeting EGFR such as Nimotuzumab and tyrosine kinase inhibitors are markedly effective on COVID-19. However, these data might raise a provisional caution regarding implication of psychiatric disorder such as schizophrenia. The author's group has been investigating the etiologic and neuropathologic associations of EGFR signaling with schizophrenia. There are significant molecular associations between schizophrenia and EGFR ligand levels in blood as well as in the brain. In addition, perinatal challenges of EGFR ligands and intraventricular administration of EGF to rodents and monkeys both resulted in severe behavioral and/or electroencephalographic endophenotypes relevant to this disorder. These animal models also display postpubertal abnormality in soliloquy-like self-vocalization as well as in intercortical functional connectivity. Here, we discuss neuropsychiatric implication of coronavirus infection and its interaction with the EGFR system, by searching related literatures in PubMed database as of the end of 2023.
Collapse
Affiliation(s)
- Hiroyuki Nawa
- Department of Physiological Sciences, School of Pharmaceutical SciencesWakayama Medical UniversityWakayamaJapan
| | - Masaaki Murakami
- Molecular Psychoneuroimmunology, Institute for Genetic MedicineHokkaido UniversitySapporoHokkaidoJapan
| |
Collapse
|
49
|
Cekin N, Akin S, Pinarbasi E, Doğan OH. Impact of IL-6 rs1800795 and rs1800796 polymorphisms on clinical outcomes of COVID-19: a study on severity of disease in Turkish population. Mamm Genome 2025; 36:213-229. [PMID: 39567384 DOI: 10.1007/s00335-024-10085-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024]
Abstract
Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is exacerbated by cytokine storms, leading to severe inflammation. Interleukin-6 (IL-6) plays a critical role in this process, and variations in its promoter may influence disease severity. This study aims to investigate the relationship between IL6 promoter polymorphisms rs1800795 (G > C) and rs1800796 (G > C) and the severity of COVID-19 in the Turkish population. A total of 332 participants were included: 84 control, 80 with mild COVID-19, and 168 with severe COVID-19. IL6 polymorphisms were genotyped using the restriction fragment length polymorphism (RFLP) method. The genotypes rs1800795 GC (OR = 3.00, 95% CI: 1.669-5.398, p < 0.000), CC (OR = 7.44, 95% CI: 2.899-19.131, p < 0.000), and rs1800796 GC (OR = 2.76, 95% CI: 1.603-4.761, p < 0.000), as well as the alleles rs1800795 C (OR = 3.01, p < 0.000) and rs1800796 C (OR = 1.97, p = 0.002), may be associated with the severity of COVID-19. According to the Jonckheere-Terpstra (J-T) test, the most significant trends that vary linearly with disease severity were observed for D-dimer [J-T = 15.896, Effect size = 0.68 (0.61 to 0.76), p < 0.000] and CRP [J-T = 15.389, Effect size = 0.66 (0.59 to 0.73), p < 0.000]. The distribution of clinical parameters across genotype combinations (rs1800796/rs1800795*) showed that GC/GC* and GC/CC* were linked to a higher risk of severe inflammation, clotting, and organ damage. Additionally, it has been determined that the G-C and C-C haplotypes may be associated with increased severity of COVID-19. The rs1800795 and rs1800796 polymorphisms are linked to COVID-19 severity and could help guide future treatment strategies.
Collapse
Affiliation(s)
- Nilgun Cekin
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey.
- Faculty of Medicine, Department of Medical Biochemistry, Sivas Cumhuriyet University, Sivas, Turkey.
| | - Seyda Akin
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Ergun Pinarbasi
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Okan Halef Doğan
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
50
|
Shahzamani K, Amooyi A, Karampoor S, Khanizadeh S, Farahmand M. Klotho protein: A key modulator of aging and COVID-19 severity. Int J Biol Macromol 2025; 296:139234. [PMID: 39798764 DOI: 10.1016/j.ijbiomac.2024.139234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 11/04/2024] [Accepted: 12/24/2024] [Indexed: 01/15/2025]
Abstract
The COVID-19 pandemic has drawn significant attention to factors affecting disease severity, especially in older adults. This study explores the relationship between Klotho, an anti-aging protein, and COVID-19 severity. Conducted at Tehran's Firouzgar Hospital, this case-control study involved 279 participants, assessing serum levels of Klotho, inflammatory markers (C-reactive protein (CRP), Interleukin 6 (IL-6)), and Vitamin D. The findings indicate significantly lower Klotho levels in COVID-19 patients, especially those in the ICU, which correlate with elevated inflammatory markers and reduced Vitamin D levels. This inverse relationship between Klotho levels and disease severity underscores the protein's potential modulatory role in the inflammatory response to COVID-19. The study not only highlights the importance of Klotho as a biomarker for aging and disease severity but also suggests its potential therapeutic value in managing COVID-19, offering a novel perspective on targeting aging-related pathways to mitigate the impact of the disease. These insights open new avenues for research and intervention strategies to leverage anti-aging mechanisms to combat COVID-19 and potentially other age-related diseases.
Collapse
Affiliation(s)
- Kiana Shahzamani
- Lorestan University of Medical Sciences, Islamic Republic of Iran.
| | - Atefeh Amooyi
- Lorestan University of Medical Sciences, Islamic Republic of Iran
| | - Sajad Karampoor
- Iran University of Medical Sciences, Islamic Republic of Iran
| | | | | |
Collapse
|