1
|
Zuo H, Yang X, Wang Y, Hu B, Zhu Z, Guo Z, Weng S, He J, Xu X. Excessive activation of JAK-STAT signaling contributes to inflammation induced by acute Vibrio infection in shrimp. Virulence 2025; 16:2451169. [PMID: 39819453 PMCID: PMC11749392 DOI: 10.1080/21505594.2025.2451169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 12/12/2024] [Accepted: 01/05/2025] [Indexed: 01/19/2025] Open
Abstract
Uncontrolled immune responses resulting from overactivated cellular signaling pathways, leading to inflammation and tissue injury, are a major cause of death in pathogen-infected individuals. This phenomenon has been well studied in mammals but is less explored in invertebrates. Bacteria of the genus Vibrio are among the most harmful pathogens to humans and aquatic animals. In shrimp, Vibrio infection is generally characterized by the sudden onset of disease, with pathological signs of opaque and whitish muscle tissue. The current study shows that shrimp acutely infected with high dose of Vibrio parahaemolyticus develop inflammation-like pathological changes, leading to rapid death. Excessive activation of JAK-STAT signaling, rather than the Dorsal and Relish pathways, results in overactivation of shrimp immunity and is a major cause of inflammation induced by acute Vibrio infection. Weakening JAK-STAT signaling attenuates the inflammatory response and reduces mortality caused by acute Vibrio infection in shrimp, whereas enhancing JAK-STAT signaling can convert a normal infection into an acute one, accelerating shrimp death. Therefore, this study indicates that, similar to that in mammals, the pathogenesis of infectious diseases in invertebrates is complicated by inflammatory responses triggered by dysregulated immune signaling.
Collapse
Affiliation(s)
- Hongliang Zuo
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Provincial Observation and Research Station for Marine Ranching in Lingdingyang bay, Zhuhai, P. R. China
| | - Xiya Yang
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Youxi Wang
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Bangping Hu
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhiming Zhu
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Provincial Observation and Research Station for Marine Ranching in Lingdingyang bay, Zhuhai, P. R. China
| | - Zhixun Guo
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Fisheries Research Institute (CAFS), Guangzhou, P. R. China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiaopeng Xu
- State Key Laboratory of Biocontrol, School of Life Sciences/Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)/China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
2
|
Kakavand G, Arabzadeh S, Mohebbi S, Saeedfar K, Abedini A, Mardani M. Impact of remdesivir treatment on factor VIII gene expression and hematological parameters in COVID-19 patients. Microb Pathog 2025; 204:107536. [PMID: 40187577 DOI: 10.1016/j.micpath.2025.107536] [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: 09/28/2024] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The novel coronavirus, COVID-19, which was first identified in December 2019 rapidly spread worldwide and was declared a global pandemic. Beyond respiratory symptoms, COVID-19 often results in coagulation and vascular endothelium disorders, causing increased clotting and bleeding, which are closely linked to the acute phase of the infection. Factor VIII is a crucial protein in the blood coagulation cascade, and elevated FVIII levels have been linked to thrombotic events in COVID-19, highlighting the need to understand its behavior during treatment. Remdesivir is an antiviral drug that has shown promise in reducing recovery time and mortality rates in COVID-19 patients. This study aims to examine the changes in blood factors and the expression of the factor VIII gene in patients treated with Remdesivir. Blood samples were collected from 30 COVID-19 patients before and after Remdesivir treatment and from 20 healthy individuals. Patients with underlying diseases were excluded from the study. RNA was extracted from these samples, followed by cDNA synthesis. The expression of the factor VIII gene was analyzed using Real-Time PCR. The results indicated that blood factors such as Urea, ALK, AST, WBC, and CRP were elevated in the patient group compared to the control group. At the same time, FBS, Urea, ALK, AST, WBC, RDW, INR, and K levels increased in the Remdesivir treatment group (P < 0.001). Conversely, MCHC, RBC, and Ca levels decreased in both patient and treatment groups compared to the control group (P < 0.001). The expression of the FVIII gene was upregulated approaching 2 times in COVID-19 patients and 1.5-fold in the treatment group compared to the control group (P < 0.001). However, no significant changes were observed in FVIII expression before and after Remdesivir treatment. However, a positive correlation between RBC, FBS, and Urea in the patient group and a negative correlation between RDW and FVIII expression levels was observed. In the treatment group, FVIII expression level correlated negatively with Urea, P, and RDW. These findings suggest that elevated FVIII levels are associated with disease severity and excessive coagulation in COVID-19 patients. Additionally, Remdesivir does not appear to exacerbate the coagulation process.
Collapse
Affiliation(s)
- Ghazal Kakavand
- Department of Biology, Faculty of Basic Science, Ale Taha Institute of Higher Education, Tehran, Iran
| | - Somayeh Arabzadeh
- Department of Biology, Faculty of Basic Science, Ale Taha Institute of Higher Education, Tehran, Iran
| | - Sohameh Mohebbi
- Department of Biology, Faculty of Basic Science, Ale Taha Institute of Higher Education, Tehran, Iran.
| | - Kayvan Saeedfar
- Tracheal Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Atefeh Abedini
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Mardani
- Shahid Beheshti University of Medical Sciences, Infectious Disease Department, Loghman Hakim Hospital, Tehran, Iran
| |
Collapse
|
3
|
Elboraay T, Ebada MA, Elsayed M, Aboeldahab HA, Salamah HM, Rageh O, Elmallahy M, AboElfarh HE, Mansour LS, Nabil Y, Eltawab AKA, Atwan H, Alkanj S. Long-term neurological and cognitive impact of COVID-19: a systematic review and meta-analysis in over 4 million patients. BMC Neurol 2025; 25:250. [PMID: 40514644 PMCID: PMC12166599 DOI: 10.1186/s12883-025-04174-9] [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: 01/18/2025] [Accepted: 04/02/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Neuropsychiatric symptoms emerged early in the COVID-19 pandemic as a key feature of the virus, with research confirming a range of neuropsychiatric manifestations linked to acute SARS-CoV-2 infection. However, the persistence of neurological symptoms in the post-acute and chronic phases remains unclear. This meta-analysis assesses the long-term neurological effects of COVID-19 in recovered patients, providing insights for mental health service planning. METHODS A comprehensive literature search was conducted across five electronic databases: PubMed, Scopus, Web of Science, EBSCO, and CENTRAL, up to March 22, 2024. Studies evaluating the prevalence of long-term neurological symptoms in COVID-19 survivors with at least six months of follow-up were included. Pooled prevalence estimates, subgroup analyses, and meta-regression were performed, and publication bias was assessed. RESULTS The prevalence rates for the different symptoms were as follows: fatigue 43.3% (95% CI [36.1-50.9%]), memory disorders 27.8% (95% CI [20.1-37.1%]), cognitive impairment 27.1% (95% CI [20.4-34.9%]), sleep disorders 24.4% (95% CI [18.1-32.1%]), concentration impairment 23.8% (95% CI [17.2-31.9%]), headache 20.3% (95% CI [15-26.9%]), dizziness 16% (95% CI [9.5-25.7%]), stress 15.9% (95% CI [10.2-24%]), depression 14.0% (95% CI [10.1-19.2%]), anxiety 13.2% (95% CI [9.6-17.9%]), and migraine 13% (95% CI [2.2-49.8%]). Significant heterogeneity was observed across all symptoms. Meta-regression analysis showed higher stress, fatigue, and headache in females, and increased stress and concentration impairment with higher BMI. CONCLUSIONS Neurological symptoms are common and persistent in COVID-19 survivors. This meta-analysis highlights the significant burden these symptoms place on individuals, emphasizing the need for well-resourced multidisciplinary healthcare services to support post-COVID recovery. REGISTRATION AND PROTOCOL This meta-analysis was registered in PROSPERO with registration number CRD42024576237.
Collapse
Affiliation(s)
- Toka Elboraay
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Mahmoud A Ebada
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt.
- Egyptian Fellowship of Neurology, Ministry of Health and Population, Cairo, Egypt.
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA.
| | - Maged Elsayed
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Heba Ahmed Aboeldahab
- Clinical Research Department, El-Gomhoria General Hospital, MOHP, Karmouz, Alexandria, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Hazem Mohamed Salamah
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Omar Rageh
- Faculty of Medicine, Tanta University, Tanta, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Mohamed Elmallahy
- Faculty of Medicine, Tanta University, Tanta, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Hadeer Elsaeed AboElfarh
- Neurology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Lena Said Mansour
- Faculty of Medicine and Internal Medicine Resident, Damanhour Teaching Hospital, Damanhour, Beheira, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Yehia Nabil
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Ahmed Khaled Abd Eltawab
- Sednawy Health Insurance Hospital, Cairo, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Hany Atwan
- Faculty of Medicine, Assiut University, Assiut, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Souad Alkanj
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| |
Collapse
|
4
|
Kasem Ali Sliman R, Cohen H, Shehadeh S, Batcir R, Alter YE, Cohen K, Koren I, Halabi I, Sliman H, Saied MH. Pediatric autoimmune diseases in the light of COVID-19 pandemic, A retrospective observational big data study. J Transl Autoimmun 2025; 10:100281. [PMID: 40162434 PMCID: PMC11951201 DOI: 10.1016/j.jtauto.2025.100281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 04/02/2025] Open
Abstract
Background The COVID-19 pandemic has raised concerns about potential links between SARS-CoV-2 infection and autoimmune diseases. This study investigated changes in the incidence rate (IR) of autoimmune diseases among children following the pandemic's onset. Methods A retrospective cross-sectional study analyzed data from Clalit Health Services, Israel's largest healthcare provider, examining the IR of different autoimmune diseases in children aged 0-18. The study compared pre-pandemic (2019) with pandemic/post-pandemic periods (2020-2023), encompassing a cohort of over 1.5 million children. Results Significant IR increases were observed across multiple autoimmune diseases. Rheumatic diseases (Juvenile Idiopathic Arthritis, Systemic Lupus Erythematosus, Henoch Schoenlein Purpura (HSP)) showed consistent increases, with HSP demonstrating the most pronounced trend. Endocrine disorders exhibited diverse patterns, with autoimmune thyroid diseases and Type 1 diabetes showing overall increases, while diabetic ketoacidosis exhibited an initial spike followed by a decline. Gastrointestinal diseases displayed heterogeneous patterns; Celiac disease and Ulcerative colitis showed general increases, Crohn's disease showed a downward trend, and autoimmune hepatitis exhibited an initial significant decrease followed by a significant increase. Dermatological conditions, including Psoriasis and Vitiligo, demonstrated consistent elevations throughout 2020-2023. Immune Thrombocytopenia Purpura showed initial decreases followed by significant increases in 2022-2023. Conclusions This comprehensive analysis reveals significant changes in pediatric autoimmune disease incidence following the COVID-19 pandemic, suggesting potential associations between SARS-CoV-2 infection and autoimmune dysregulation. The diverse patterns observed across different conditions highlight the complex interplay between viral infection and autoimmunity, emphasizing the need for continued surveillance and investigation of long-term immunological consequences of COVID-19 in pediatric populations.
Collapse
Affiliation(s)
- Rim Kasem Ali Sliman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
| | - Hilla Cohen
- Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
| | - Shereen Shehadeh
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Infectious Disease Unit, Carmel Medical Center, Haifa, Israel
| | - Reut Batcir
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Gastroenterology Unit, Carmel Medical Center, Haifa, Israel
| | - Yigal Elenberg Alter
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Gastroenterology Unit, Carmel Medical Center, Haifa, Israel
| | - Keren Cohen
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Ilana Koren
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Inbal Halabi
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Hussein Sliman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Cardiology, Carmel Medical Center, Heart Center, Haifa, Israel
| | - Mohamad Hamad Saied
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Center, Utrecht, the Netherlands
| |
Collapse
|
5
|
Amrouche T, Lammi S, Drider D. Probiotics and Prebiotics Intervention in Respiratory and Digestive Infections Linked to Covid-19. Probiotics Antimicrob Proteins 2025; 17:1356-1367. [PMID: 39614066 DOI: 10.1007/s12602-024-10404-2] [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] [Accepted: 11/11/2024] [Indexed: 12/01/2024]
Abstract
Probiotics and prebiotics have been suggested as natural agents against viral infections and dysbiosis and may encourage clinical applications. This review aims to analyze the main and recent advances related to viral infections such as Covid-19 and its gastrointestinal complications, antiviral immunity generated and possible preventive role that probiotics and/or prebiotics can play in controlling and promoting antiviral immunity. The literature search was performed through a critical analysis of relevant publications reported in PubMed and Scopus databases on clinical trials and assays conducted in vitro on colon cells and in vivo on mice. Some studies using probiotics and prebiotics for the prevention of viral infection in different age groups are discussed. Covid-19 patients have been shown to suffer from gastrointestinal complications in addition to respiratory symptoms due to interactions between the respiratory system and the gastrointestinal tract infected with SARS-CoV-2. Unfortunately, therapies used to prevent (or treat) symptoms of Covid-19 have proven to be of limited effectiveness. In addition, the lack of access to coronavirus vaccines around the world and vaccine hesitancy continue to hamper control of Covid-19. It is therefore crucial to find alternative methods that can prevent disease symptoms. Evidence-based efficacy of certain probiotics (Lactobacillus and Bifidobacterium) that may be useful in viral infections was shown with immunomodulatory properties (pro-inflammatory mediators reduction), promoting antiviral immunity (antibodies production, virus titers) and controlling inflammation (anti-inflammatory effect), as well as viral clearance and antimicrobial potential against opportunistic bacteria (anti-dysbiosis effect). But, available data about clinical application of probiotics in Covid-19 context remain limited and relevant scientific investigation is still in its early stages. Also, evidence for prebiotics potential in this field is limited, since the exact mechanism involved in systemic immune modulation by these compounds is till now unknown. Thus, further research is necessary to explore in the viral infection context the mechanism by which gut and lung interact in the presence of probiotics and prebiotics through more animal and clinical experiments.
Collapse
Affiliation(s)
- Tahar Amrouche
- Laboratoire Qualité Et Sécurité Des Aliments, Faculté Des Sciences Biologiques Et Des Sciences Agronomiques, Université Mouloud Mammeri, 15 000, Tizi Ouzou, Algeria.
| | - Sarah Lammi
- Laboratoire Qualité Et Sécurité Des Aliments, Faculté Des Sciences Biologiques Et Des Sciences Agronomiques, Université Mouloud Mammeri, 15 000, Tizi Ouzou, Algeria
| | - Djamel Drider
- UMR Transfrontalière BioEcoAgro INRAE 1158, Université de Lille (ULille), 59000, Lille, France
| |
Collapse
|
6
|
Siciliani L, Cappa G, Zattera C, Albi G, Mondelli MU, Marzi L. Altered liver hemodynamics in patients with COVID-19: a cross sectional study. J Ultrasound 2025; 28:437-445. [PMID: 40172816 PMCID: PMC12145364 DOI: 10.1007/s40477-025-01012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/16/2025] [Indexed: 04/04/2025] Open
Abstract
AIMS Abnormalities in liver biochemistry are common in COVID-19 patients. Hepatic vein Doppler waveform, typically triphasic, may become biphasic or monophasic in cirrhosis, correlating with liver dysfunction, fibrosis, inflammation, and portal hypertension. This study investigates liver ultrasound (US) features in COVID-19 patients, correlating hepatic vein Doppler waveform and portal vein velocity (PVV) with inflammatory indexes and clinical outcomes. METHODS Fifty-seven patients with SARS-CoV-2 infection participated in a crosssectional study. Bedside upper abdomen US evaluations, including B-mode and Doppler, were conducted using a convex probe. Hepatic vein Doppler waveforms were classified as triphasic, biphasic, or monophasic, and the hepatic vein waveform index (HVWI) was calculated. PVV was measured over three cardiac cycles. Tracings were blindly analyzed by three operators to ensure consistency. RESULTS Low HVWI and high PVV correlated with elevated LDH, ALT, D-dimer, and ferritin (p < 0.05). HVWI showed significant negative correlations with ferritin, D-dimer, and ALT (p < 0.05). D-Dimer and ferritin were higher in patients with biphasic/monophasic waveforms (p < 0.05). High PVV and larger spleen diameters predicted worse respiratory outcomes, including CPAP and tracheal intubation (p < 0.05). Optimal cut-off values for PVV (21.7 cm/s) and spleen diameter (9.84 cm) maximized sensitivity and specificity for predicting these outcomes. FIB-4 scores did not correlate with respiratory outcomes or hepatic hemodynamics (p > 0.05). Hemodynamic alterations were not significantly influenced by the presence of SLD (Steatotic Liver Disease). CONCLUSIONS COVID-19 patients exhibit altered intrahepatic hemodynamics, with hepatic vein waveform abnormalities potentially reflecting liver inflammation and fibrosis. PVV and spleen diameter may serve as non-invasive predictors of respiratory outcomes.
Collapse
Affiliation(s)
- Luisa Siciliani
- Division of Clinical Immunology and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Giovanni Cappa
- Emergency Medicine Unit and Emergency Medicine Postgraduate Training Program, IRCCS Policlinico San Matteo University Hospital, University of Pavia, Pavia, Italy
| | - Caterina Zattera
- Emergency Medicine Unit and Emergency Medicine Postgraduate Training Program, IRCCS Policlinico San Matteo University Hospital, University of Pavia, Pavia, Italy
| | - Giuseppe Albi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Mario Umberto Mondelli
- Division of Clinical Immunology and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Marzi
- Gastroenterology Department, Bolzano Regional Hospital, 39100, Bolzano, Italy
| |
Collapse
|
7
|
Yasmin S, Ansari MY. A detailed examination of coronavirus disease 2019 (COVID-19): Covering past and future perspectives. Microb Pathog 2025; 203:107398. [PMID: 39986548 DOI: 10.1016/j.micpath.2025.107398] [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: 07/06/2024] [Revised: 01/07/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
The COVID-19 disease has spread rapidly across the world within just six months, affecting 169 million people and causing 3.5 million deaths globally (2021). The most affected countries include the USA, Brazil, India, and several European countries such as the UK and Russia. Healthcare professionals face new challenges in finding better ways to manage patients and save lives. In this regard, more comprehensive research is needed, including genomic and proteomic studies, personalized medicines and the design of suitable treatments. However, finding novel molecular entities (NME) using a standard or de novo strategy to drug development is a time-consuming and costly process. Another alternate strategy is discovering new therapeutic uses for old/existing/available medications, known as drug repurposing. There are a variety of computational repurposing methodologies, and some of them have been used to counter the coronavirus disease pandemic of 2019 (COVID-19). This review article compiles recently published data on the origin, transmission, pathogenesis, diagnosis, and management of the coronavirus by drug repurposing and vaccine development approach. We have attempted to screen probable drugs in clinical trials by using literature survey. This systematic review aims to create priorities for future research of drugs repurposed and vaccine development for COVID-19.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India; Ibne Seena College of Pharmacy, Azmi Vidya Nagri Anjhi Shahabad, Hardoi, Uttar Pradesh (U.P.) 241124, India.
| |
Collapse
|
8
|
Le VT, Yuune JPT, Vu TTP, Malik MS, Ou YY. DeepCR: predicting cytokine receptor proteins through pretrained language models and deep learning networks. J Biomol Struct Dyn 2025:1-18. [PMID: 40448687 DOI: 10.1080/07391102.2025.2512448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 05/21/2025] [Indexed: 06/02/2025]
Abstract
Cytokine receptors play a pivotal role in mediating the immune response and are critical in cytokine storms, which underlie the pathogenesis of conditions such as acute respiratory distress syndrome (ARDS) and autoimmune disorders. Identifying cytokine receptors is essential for understanding their biological functions, exploring therapeutic targets, and guiding clinical interventions. Traditional biochemical methods to identify cytokine receptors are labor-intensive, costly, and time-consuming, prompting the need for more efficient alternatives. Recent advances in computational biology have enabled the use of machine learning to classify cytokine receptor proteins. Most existing approaches focused on homologous features and protein composition to classify cytokine families, but no dedicated studies have been conducted on cytokine receptor proteins. This gap presents an opportunity to develop a method specifically for classifying cytokine receptors among other membrane proteins. In this study, we present a novel classification framework combining pre-trained language models (PLMs) with a multi-window convolutional neural network (mCNN) architecture for the fast and accurate identification of cytokine receptor proteins. PLMs, such as ProtTrans and ESM variants, capture biochemical context directly from raw protein sequences, while mCNN efficiently extracts local and global sequence patterns using convolutional layers with varying window sizes. Our model achieved an AUC of 0.96 in the training as well as 0.97 and 0.93 in two independent tests, demonstrating its effectiveness in distinguishing cytokine receptors from non-cytokine receptor proteins. By eliminating the need for manual feature extraction, this approach offers a robust and scalable solution for protein classification, paving the way for its application in drug discovery and understanding cytokine-mediated diseases.
Collapse
Affiliation(s)
- Van The Le
- Department of Computer Science and Engineering, Yuan Ze University, Chung-Li, Taiwan
| | | | - Thi Thu Phuong Vu
- Graduate Program in Biomedical Informatics, Yuan Ze University, Chung-Li, Taiwan
| | - Muhammad Shahid Malik
- Department of Computer Science and Engineering, Yuan Ze University, Chung-Li, Taiwan
- Department of Computer Sciences, Karakoram International University, Gilgit-Baltistan, Pakistan
| | - Yu-Yen Ou
- Department of Computer Science and Engineering, Yuan Ze University, Chung-Li, Taiwan
- Graduate Program in Biomedical Informatics, Yuan Ze University, Chung-Li, Taiwan
| |
Collapse
|
9
|
Kumar N, Segovia D, Kumar P, Atti HB, Kumar S, Mishra J. Mucosal implications of oral Jak3-targeted drugs in COVID patients. Mol Med 2025; 31:203. [PMID: 40410684 PMCID: PMC12100796 DOI: 10.1186/s10020-025-01260-z] [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: 03/18/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
The JAK family, particularly JAK3, plays a crucial role in immune signaling and inflammatory responses. Dysregulated JAK3 activation in SARS-CoV-2 infections has been associated with severe inflammation and respiratory complications, making JAK inhibitors a viable therapeutic option. However, their use raises concerns regarding immunosuppression, which could increase susceptibility to secondary infections. While long-term adverse effects are less of a concern in acute COVID-19 treatment, patient selection and monitoring remain critical. Furthermore, adverse effects associated with oral JAK3 inhibitors necessitate the exploration of alternative strategies to optimize therapeutic efficacy while minimizing risks. This review highlights the role of JAK3 in immune and epithelial cells, examines the adverse effects of oral JAK3 inhibitors in COVID-19 and other treatments, and discusses alternative therapeutic strategies for improving patient outcomes.
Collapse
Affiliation(s)
- Narendra Kumar
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA.
| | - Daniel Segovia
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA
| | - Priyam Kumar
- University of Pennsylvania, Philadelphia, PA, USA
| | - Hima Bindu Atti
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA
| | - Soaham Kumar
- Veterans Memorial High School, Corpus Christi, TX, USA
| | - Jayshree Mishra
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA.
| |
Collapse
|
10
|
Raphael J, Le B, Singh S, Blanchette P, Trudeau M, Lam M, Cheung M. Early mortality in patients with cancer and COVID-19 infection treated with immunotherapy. BMC Cancer 2025; 25:922. [PMID: 40405109 PMCID: PMC12100880 DOI: 10.1186/s12885-025-14318-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Immunotherapy in the presence of COVID-19 infections raises concerns because of potential overlapping clinical complications and immune system enhancement. Further investigation is warranted to establish its safety and to improve clinical decisions. METHODS We conducted a retrospective cohort study using linked health administrative data from Ontario, Canada to assess 30-day mortality in patients with solid tumors who were treated with immunotherapy within 120 days before testing positive for COVID-19. A stepwise multivariable logistic regression model was used to identify clinical factors associated with 30-day mortality. RESULTS Between January 2020 and April 2023, 281 patients tested positive for COVID-19 and were included in our study. The mean age was 68 (Standard Deviation: 10.3), 45% (127/281) were females and 58% (163/281) had lung cancer. 59% of patients (167/281) were treated with single agent immunotherapy, and almost 80% received at least one dose of COVID-19 vaccine. The 30-day mortality was 22% (63/281) and < 5% of patients were admitted to ICU or required ventilation. Factors associated with higher mortality were older age (Odds Ratio (OR) 1.60, 95% confidence interval (CI) 1.07-2.39), prior radiation therapy (OR 2.38, 95%CI 1.08-5.28), lower hemoglobin (< 10 g/dl) (OR 4.08, 95%CI 1.89-8.82) and higher leucocytes count (> 11,000/mm3) (OR 3.63, 95%CI 1.55-8.52). CONCLUSIONS Immunotherapy does not seem to increase the risk of 30-day mortality in patients with COVID-19 infections compared to published outcomes of patients with cancer and COVID-19. Mortality was associated with certain clinical characteristics that need to be carefully examined when prescribing immunotherapy during future comparable pandemics.
Collapse
Affiliation(s)
- Jacques Raphael
- Division of Medical Oncology, Department of Oncology, Verspeeten Family Cancer Centre, Western University, 800 Commissioners Road East, London, ON, N6A 5W9, Canada.
- ICES Western, London, ON, Canada.
| | | | - Simron Singh
- Division of Medical Oncology & Hematology, Sunnybrook Odette Cancer Centre, Toronto, ON, Canada
| | - Phillip Blanchette
- Division of Medical Oncology, Department of Oncology, Verspeeten Family Cancer Centre, Western University, 800 Commissioners Road East, London, ON, N6A 5W9, Canada
- ICES Western, London, ON, Canada
| | - Maureen Trudeau
- Division of Medical Oncology & Hematology, Sunnybrook Odette Cancer Centre, Toronto, ON, Canada
| | | | - Matthew Cheung
- Division of Medical Oncology & Hematology, Sunnybrook Odette Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
11
|
Sun J, Zhou L, Li Z, He G, Mao H, Zhao J, Hunt JA, Chen X. Perovskite-Graphene Heterostructure Biosensor Integrated with Biotunable Nanoplasmonic Ternary Logic Gate for Ultrasensitive Cytokine Detection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e03124. [PMID: 40397015 DOI: 10.1002/advs.202503124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 05/01/2025] [Indexed: 05/22/2025]
Abstract
The integration of 2D-materials and optoelectronic devices has attracted great attention for advanced applications. We propose the first perovskite/graphene heterostructure-based FET biosensor with uniquely biotunable ternary logic gating functionality. The biosensor integrates a lateral perovskite-on-graphene heterostructure phototransistor with a vertical bio-nano-photonic filter, with a decoupled construction inset. In the phototransistor, photoactive perovskite quantum dots (PQDs) serve as sensitizers to absorb light while a high mobility single-layer graphene (SLG) acts as an expressway for carrier transport. In the bio-nano-photonic filter, a localized surface plasmon resonance (LSPR) is induced by gold nanoparticles (AuNPs) in conjunction with antigen-antibody binding, tuning the delivery of light passing through the filter and facilitating biotunable functionality with ternary modes. The biosensor is set up to detect human interleukin-6 (IL6) in order to determine and achieve ultrahigh sensitivity with a limit of detection (LOD) of 0.9 fg mL-1 (43 aM), which is 4 orders of magnitude greater than graphene-FET biosensors. This ultrahigh sensitivity is achieved due to the synergistic effect of PQDs/SLG heterostructure, exhibiting superior electrical, optical, and physicochemical properties, consequently providing significantly high performance of the biosensor in terms of label-free, ultrahigh sensitivity (attomolar level), rapid responsivity (5 min), excellent stability, and selectivity. This heterostructure-based biotunable configuration could open a new avenue for 2D materials in the realm of next-generation bio-nano-photonic platforms for applications in healthcare, early diagnosis, and rapid detection.
Collapse
Affiliation(s)
- Jiaxing Sun
- School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Lin Zhou
- Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Zening Li
- Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Guolin He
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Hongju Mao
- Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Jianlong Zhao
- Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - John A Hunt
- Medical Technologies Innovation Facility, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Xianfeng Chen
- School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| |
Collapse
|
12
|
Ghosh S, Das Sarma J. The age-dependent neuroglial interaction with peripheral immune cells in coronavirus-induced neuroinflammation with a special emphasis on COVID-19. Biogerontology 2025; 26:111. [PMID: 40380990 DOI: 10.1007/s10522-025-10252-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
Neurodegenerative diseases are chronic progressive disorders that impair memory, cognition, and motor functions, leading to conditions such as dementia, muscle weakness, and speech difficulties. Aging disrupts the stringent balance between pro- and anti-inflammatory cytokines, increasing neuroinflammation, which contributes to neurodegenerative diseases. The aging brain is particularly vulnerable to infections due to a weakened and compromised immune response and impaired integrity of the blood-brain barrier, allowing pathogens like viruses to trigger neurodegeneration. Coronaviruses have been linked to both acute and long-term neurological complications, including cognitive impairments, psychiatric disorders, and neuroinflammation. The virus can induce a cytokine storm, damaging the central nervous system (CNS) and worsening existing neurological conditions. Though its exact mechanism of neuroinvasion remains elusive, evidence suggests it disrupts the blood-brain barrier and triggers immune dysregulation, leading to persistent neurological sequelae in elderly individuals. This review aims to understand the interaction between the peripheral immune system and CNS glial cells in aged individuals, which is imperative in addressing coronavirus-induced neuroinflammation and concomitant neurodegeneration.
Collapse
Affiliation(s)
- Satavisha Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata, 741246, India.
- Department of Ophthalmology, University of Pennsylvania, 19104, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Li H, Zhang Y, Du S, Shen J, Liu X, Jing J. "Remodeling the intestinal immune microenvironment": immune regulation and tissue regeneration by mesenchymal stem/stromal cells in the repair microenvironment of inflammatory bowel disease. Front Immunol 2025; 16:1543702. [PMID: 40433382 PMCID: PMC12106535 DOI: 10.3389/fimmu.2025.1543702] [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: 12/11/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
The global prevalence of inflammatory bowel disease (IBD) has significantly increased in recent decades. IBD is a long-term, recurring, gastrointestinal inflammatory condition that mainly comprises two primary clinical types: ulcerative colitis and Crohn's disease. The current treatment paradigm for IBD primarily focuses on symptom management. However, this approach does not support mucosal epithelial repair, maintenance of barrier homeostasis, or regulation of biological functions in the gut. Conventional therapies rely on the frequent use of high-dose medications, including antibiotics, nonsteroidal anti-inflammatory drugs, biological agents, and immunomodulators. Recently, mesenchymal stem/stromal cells (MSCs) have gained interest in tissue regeneration owing to their unique ability to differentiate and secrete regulatory factors, including extracellular vesicles (EVs), which play crucial roles in abnormal organization. Various routes of administration have been explored in preclinical and clinical studies to deliver MSCs from diverse tissue sources. The routes include intraperitoneal, intravenous, and local (intracolonic or rectal) delivery. The MSCs employed were obtained from various tissues, including bone marrow, umbilical cord, and adipose tissue. This article reviews the research framework for the application of MSCs and EVs secretion in the treatment of IBD, emphasizing key immunological effects, such as immune microenvironment regulation, intestinal barrier stabilization, and therapeutic approaches targeting intestinal barrier disorders. The discussion primarily focuses on the advantages of MSCs over other biologics, impairment of gut mucosal tissue-resident mesenchymal stem cells in IBD development, immune targets (at the cellular and molecular levels) within the framework of IBD, and the reparative effects of MSCs in the microenvironment of IBD. We aimed to present an overview of the current trends in MSC research and therapy, as well as to identify the challenges and future directions that must be addressed to advance research on MSC-mediated therapeutic strategies for IBD.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Jing
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
14
|
Gao X, Shi L, Jing D, Ma C, Wang Q, Wang J, Zhu F, Zhao M, Chen Y, Zhou G. A Rare Case of Small Bowel Ulceration Induced by COVID-19. J Inflamm Res 2025; 18:6123-6131. [PMID: 40376594 PMCID: PMC12079040 DOI: 10.2147/jir.s507209] [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: 11/18/2024] [Accepted: 04/12/2025] [Indexed: 05/18/2025] Open
Abstract
Background COVID-19 can affect multiple organ systems beyond the respiratory tract, including the gastrointestinal tract, where gastrointestinal symptoms include nausea, vomiting, diarrhea, abdominal pain, and even serious manifestations such as ulcers, perforation, or gastrointestinal bleeding. Case Presentation We report a case of a 45-year-old male patient with small bowel ulcers caused by chronic COVID-19 infection. Initially presenting with fever and transient unconsciousness, he developed ischemic necrosis and required a mid-thigh amputation. Despite treatment with anti-infection therapy, extracorporeal membrane oxygenation, and continuous renal replacement therapy, he experienced persistent abdominal pain and gastrointestinal bleeding. Imaging and colonoscopy confirmed partial small bowel obstruction and inflammation. After treatment with methylprednisolone and enteral nutrition, his symptoms improved. However, he suffered a gastrointestinal perforation requiring emergency surgery and later underwent a successful stoma reversal. The patient was subsequently discharged with improvement and was discharged with a primary diagnosis of "enterostomal status, perforation of small intestinal ulcer, viral myocarditis, COVID-19 infection, and post right lower extremity amputation". During the past year of follow-up, the patient has not experienced any recurrence of abdominal pain or rectal bleeding. Conclusion Although coronavirus pneumonia combined with small bowel ulcers is rare, it requires emergency treatment and has a high mortality rate. This case highlighted the severe gastrointestinal complications induced by COVID-19 infection and the effectiveness of comprehensive management strategies.
Collapse
Affiliation(s)
- Xizhuang Gao
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Lihao Shi
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, People’s Republic of China
| | - Dehuai Jing
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Cuimei Ma
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Quanyi Wang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Jiehuan Wang
- Department of Imaging, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Mengmeng Zhao
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Yun Chen
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, People’s Republic of China
| |
Collapse
|
15
|
Li X, Pakanati V, Liu C, Wang T, Morelli D, Korpak A, Baraff A, Isaacs SN, Vittor A, Chang KM, Le E, Smith NL, Lee JS, Ross JM, Shah JA. Peripheral blood cytokine profiles predict the severity of SARS-CoV-2 infection: an EPIC 3 study analysis. BMC Infect Dis 2025; 25:677. [PMID: 40340594 PMCID: PMC12063214 DOI: 10.1186/s12879-025-10914-6] [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: 10/08/2024] [Accepted: 04/03/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Predicting which patients will develop severe COVID-19 complications could improve clinical care. Peripheral blood cytokine profiles may predict the severity of SARS-CoV-2 infection, but none have been identified in US Veterans. METHODS We analyzed peripheral blood cytokine profiles from 202 participants in the EPIC3 study, a prospective observational cohort of US Veterans tested for SARS-CoV-2 across 15 VA medical centers. Illness severity was assessed based on the highest level documented during the first 60 days after recruitment. We correlated cytokine levels with illness severity using LASSO logistic regression, random forest, and XGBoost models on a 70% training set and calculated the AUC on a 30% test set. RESULTS LASSO regression identified 6 cytokines as predictors of SARS-CoV-2 severity with 77.3% AUC in the test set. Random forest and XGBoost models achieved an AUC of 80.4% and 80.7% in the test set, respectively. All models assigned a feature importance to each cytokine, with IP-10, MCP-1, and HGF consistently identified as key markers. CONCLUSIONS Cytokine profiles are predictive of SARS-CoV-2 severity in US Veterans and may guide tailored interventions for improved patient management.
Collapse
Affiliation(s)
- Xumin Li
- VA Puget Sound Health Care System, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | | - Cindy Liu
- VA Puget Sound Health Care System, Seattle, WA, USA
| | - Tracy Wang
- VA Puget Sound Health Care System, Seattle, WA, USA
| | | | - Anna Korpak
- VA Puget Sound Health Care System, Seattle, WA, USA
| | - Aaron Baraff
- VA Puget Sound Health Care System, Seattle, WA, USA
| | - Stuart N Isaacs
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Vittor
- North Florida/South Georgia Veterans Health System, Gainesville, FL, USA
- Division of Infectious Disease and Global Medicine, University of Florida, Gainesville, FL, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Le
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Nicholas L Smith
- VA Puget Sound Health Care System, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jennifer S Lee
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University, Palo Alto, CA, USA
| | - Jennifer M Ross
- VA Puget Sound Health Care System, Seattle, WA, USA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Javeed A Shah
- VA Puget Sound Health Care System, Seattle, WA, USA.
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Anzic N, Stoiber W, Obermeyer A, Mertz KD, Stalder A, Haslbauer JD, Tzankov A. Two-sided effects of neutrophil extracellular traps and changes in the myeloid compartment in acute COVID-19: A histopathological study on autopsy cases from the first and second COVID-19 waves in Switzerland. J Leukoc Biol 2025; 117:qiaf056. [PMID: 40356380 DOI: 10.1093/jleuko/qiaf056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/05/2025] [Accepted: 05/12/2025] [Indexed: 05/15/2025] Open
Abstract
Severe COVID-19 is characterized by complex immunopathology that involves inflammation, endothelial dysfunction, and immunothrombosis. Neutrophil extracellular traps (NETs) have been recognized as key factors in the severity of the disease, with their emergence correlating to viral load, immmunothrombosis, and organ damage. In this study, we investigated the role of NETosis and macrophage activation in the course of severe COVID-19. We analyzed 23 autopsy samples from patients who died from COVID-19 and performed immunohistochemical staining and stereological point counting to quantify leukocyte infiltration and NET formation among other histopathological parameters. Our results showcase 2 evident immunophenotypes: lowNET and highNET. The lowNET group displayed lower NET formation, higher viral loads, and an increased incidence of secondary infections, as well as shorter survival times. In contrast, the highNET group exhibited increased neutrophil activation, pronounced endothelial damage and thrombotic complications, as well as prolonged survival times. Our data suggest a dual role of NETosis in COVID-19: initially protective, limiting viral replication, but later likely detrimental through immunothrombosis and tissue damage. These findings underline the need for tailored therapeutic actions, with early antiviral and immune-modulating interventions for lowNET patients and strategies aiming to limit excessive NETosis and coagulopathy in highNET patients. Further research is needed to define the timing of interventions based on the dynamics of NETosis.
Collapse
Affiliation(s)
- Nina Anzic
- Institute of Pathology, Cantonal Hospital Luzern, Spitalstrasse 16, 6000 Luzern, Switzerland
| | - Walter Stoiber
- Department of Environment and Biodiversity, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Astrid Obermeyer
- Department of Environment and Biodiversity, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Kirsten D Mertz
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Anna Stalder
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Jasmin D Haslbauer
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| |
Collapse
|
17
|
Shujath J. Beyond Traditional Publishing: Social Media as a Catalyst for Biomedical Research Dissemination and Collaboration. J Interferon Cytokine Res 2025. [PMID: 40329892 DOI: 10.1089/jir.2025.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
The expansion of social media has fundamentally transformed biomedical research dissemination and collaboration, particularly within the interferon and cytokine research community. This paper explores recent trends (2024-2025) that have amplified the role of platforms such as Twitter (now "X"), LinkedIn, Mastodon, Threads, and Bluesky. These tools have facilitated rapid knowledge exchange, democratized access to scientific discourse, enabled diverse voices to participate meaningfully, and fostered cross-disciplinary and global collaborations. Additionally, the integration of preprint repositories like bioRxiv and medRxiv, along with the evolution of open access publishing, further accelerates the accessibility and immediacy of scientific communication. Despite evident benefits, the rapid dissemination facilitated by social media also poses ethical challenges, including concerns about misinformation, premature dissemination of preliminary data, and privacy considerations. Practical strategies for researchers and institutions to effectively navigate these platforms responsibly are presented, aiming to optimize the impact of social media on scientific discovery and public engagement.
Collapse
Affiliation(s)
- Jaleel Shujath
- University of the District of Columbia, Washington, District of Columbia, USA
| |
Collapse
|
18
|
Yang L, Zhou X, Liu J, Yang G, Tan W, Ding H, Fang X, Yu J, Li W, He J, Cao H, Ma Q, Yu L, Lu Z. PEBL, a component-based Chinese medicine, reduces virus-induced acute lung injury by targeting FXR to decrease ACE2 levels. J Adv Res 2025:S2090-1232(25)00295-4. [PMID: 40324631 DOI: 10.1016/j.jare.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/16/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION Despite the growing clinical need, the therapeutic efficacy of drugs for acute lung injury (ALI) remains inadequate. Traditional Chinese Medicine (TCM) holds potential in managing ALI due to its unique therapeutic properties. However, the intricate nature of TCM formulations hinders global adoption. Component-based Chinese medicine (CCM) offers a promising pathway for TCM's internationalization. Phillyrin-Emodin-Baicalin-Liquiritin (PEBL), a CCM with significant anti-inflammatory activity, is derived from the well-established TCM formula Liang-Ge-San. Whether PEBL effectively addresses viral ALI, however, remains unclear. OBJECTIVES This study aims to investigate the therapeutic effects and underlying mechanisms of PEBL on viral ALI. METHODS The efficacy of PEBL against Poly(I:C)-induced ALI was assessed by analyzing cytokine production, macrophage infiltration, pulmonary damage, and mortality. Bioinformatics and network pharmacology were employed to identify key targets and signaling pathways. The molecular mechanisms were further validated using Poly(I:C)-treated RAW264.7 cells, Tg(coro1α: GFP) zebrafish, BALB/c mice, and models of Influenza A/Puerto Rico/8/1934 (H1N1) virus strain (PR8)-induced ALI in BALB/c mice and SARS-CoV-2 Omicron XBB.1.16 subvariant (XBB)-induced ALI in hACE2-transgenic C57BL/6 mice. RESULTS PEBL mitigated Poly(I:C)-induced ALI, as evidenced by reduced cytokine levels, diminished macrophage infiltration, alleviated lung damage, and decreased mortality. Virtual screening identified the farnesyl X receptor (FXR) and angiotensin-converting enzyme 2 (ACE2) as key therapeutic targets for viral pneumonia. Mechanistically, PEBL downregulated FXR expression, inhibiting FXR binding to ACE2 promoters, which subsequently suppressed NF-κB-p65 nuclear translocation and cytokine production. In vivo, PEBL attenuated cytokine production by inhibiting ACE2 transcription through FXR downregulation, leading to alleviation of Poly(I:C)-induced ALI in both zebrafish and mice. Additionally, PEBL significantly improved symptoms of ALI caused by PR8 and XBB infections, by disrupting the FXR/ACE2 signaling axis, resulting in reduced weight loss, lower lung indices, diminished viral load and titer, fewer pulmonary lesions, and suppressed NF-κB-p65 nuclear translocation, along with decreased cytokine storm. CONCLUSIONS This study provides the first evidence that PEBL offers protective effects against ALI induced by acute respiratory viruses. PEBL prevents FXR from binding to ACE2 by inhibiting FXR transcription, which reduces macrophage infiltration, cytokine storm formation, and inflammatory injury, thereby ameliorating viral ALI. These findings underscore the potential of PEBL as a candidate for further exploration in the treatment of viral ALI.
Collapse
Affiliation(s)
- Liling Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Pharmacy, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; International Joint Laboratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Southern Medical University, Guangzhou 510030, China
| | - Guangli Yang
- Department of Central Laboratory, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Weifu Tan
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Hongyan Ding
- Omega-3 Research and Conversion Center, Dongguan Innovation Research Institute, Guangdong Medical University, Dongguan 523808, China
| | - Xiaochuan Fang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Wei Li
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Jiayang He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, China
| | - Huihui Cao
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
19
|
Valencia-Blancas T, Pérez-Orozco LH, Durán-Gómez V, García-Barboza E, López-Anguiano RR. [Atypical clinical and mortality in older adults hospitalized with COVID-19 pneumonia]. REVISTA MEDICA DEL INSTITUTO MEXICANO DEL SEGURO SOCIAL 2025; 63:e6350. [PMID: 40332445 PMCID: PMC12122060 DOI: 10.5281/zenodo.15178449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/05/2025] [Indexed: 05/08/2025]
Abstract
Background Pneumonia caused by SARS-CoV-2 is a public health problem. Older adults are vulnerable and the atypical presentation delays diagnosis and increases mortality. In this group the atypical clinical manifestations are more frequent. Objective To identify the clinical manifestations and mortality of COVID-19 pneumonia in hospitalized older adults. Material and methods A comparative cross-sectional observational study was carried out, from July 1, 2020, to December 31, 2021, in a general hospital of Mexico City. Patients aged 60 years or more who were hospitalized with a diagnosis of COVID-19 were included. Results 267 older adults participated; 53.9% were men, with a median age of 74 years. The most frequent comorbidities in this population were type 2 diabetes mellitus (T2DM), hypertension and chronic obstructive pulmonary disease (COPD). 41.2% showed atypical presentation of the disease; mortality in the studied population was 53.5%. Conclusions The atypical presentation of COVID-19 pneumonia is common in adults over 60 years of age and it increases significantly as they become older. The absence of typical symptoms of pneumonia and the ignorance of the most common signs could delay diagnosis and timely care in this age group, which may increase complications and mortality.
Collapse
Affiliation(s)
- Tlalnelli Valencia-Blancas
- Instituto Mexicano del Seguro Social, Hospital General Regional No. 2, Servicio de Geriatría. Ciudad de México, MéxicoInstituto Mexicano del Seguro SocialMéxico
| | - Lucía Herlinda Pérez-Orozco
- Instituto Mexicano del Seguro Social, Hospital General de Zona No. 27, Servicio de Geriatría. Ciudad de México, MéxicoInstituto Mexicano del Seguro SocialMéxico
| | - Verónica Durán-Gómez
- Instituto Mexicano del Seguro Social, Hospital General de Zona No. 27, Servicio de Geriatría. Ciudad de México, MéxicoInstituto Mexicano del Seguro SocialMéxico
| | - Evelín García-Barboza
- Instituto Mexicano del Seguro Social, Hospital General de Zona No. 27, Servicio de Medicina Interna. Ciudad de México, MéxicoInstituto Mexicano del Seguro SocialMéxico
| | - Roberto Rivelino López-Anguiano
- Instituto Mexicano del Seguro Social, Órgano de Operación Administrativa Desconcentrada Ciudad de México Norte, Coordinación Auxiliar Médica de Investigación en Salud. Ciudad de México, MéxicoInstituto Mexicano del Seguro SocialMéxico
| |
Collapse
|
20
|
Haider L, Rommer P, Ahmet I, Dena A, Thurnher AP, Tomassino E, Bonderman D, Thalhammer F, Seidel S, Berger T, Thurnher M. No brain MRI abnormalities after mild-to-moderate COVID-19: an observational study. Neuroradiology 2025:10.1007/s00234-025-03586-1. [PMID: 40314790 DOI: 10.1007/s00234-025-03586-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 03/05/2025] [Indexed: 05/03/2025]
Abstract
PURPOSE To assess COVID-19-related morphological brain changes in individuals who recovered from mild-to-moderate COVID-19. METHOD This prospective cohort study enrolled 112 consecutive individuals who recovered from mild-to-moderate COVID-19 and underwent an MRI of the brain between September 2020 and March 2022. MR exams were consistently obtained on a clinical 3T MR scanner in all study participants and 50 age-matched matched controls. The following clinical neuroradiological MR imaging findings were analyzed: post- and acute ischemic lesions, cortical signal alterations, microbleeds, perfusion abnormalities, cytotoxic lesions of the corpus callosum, and vascular abnormalities. Additionally, we manually quantified white matter lesion loads and the number of perivascular spaces and performed an automated brain volumetric analysis. RESULTS In 112 consecutive individuals the mean age was 45 years, female: male = 70:42, mean days at MRI after SARS CoV-2 infection: 228 (sd: 140), and hospitalized: non-hospitalized ratio = 30:82. Using general linear regression models, adjusting for age and gender, the frequency of white matter hyperintensities was not significantly different between subjects who recovered from COVID-19 and matched controls: 9.8 (sd: 17.3) vs. 7.6 (sd: 12.7), p = 0.590. Similarly, the number of enlarged perivascular spaces was not significantly different between the two groups: 62.7 (sd: 43.5) vs. 61.3 (sd: 47.2), p = 0.902. A subgroup analysis between those who were hospitalized in the course of the disease, in which no one required intensive care, and those who remained outpatients, also did not reveal any differences in MRI measures. We did not find evidence for perfusion-/diffusion abnormalities, (micro-)hemorrhages, or cortical abnormalities. CONCLUSIONS In the present cohort, there was currently no evidence of COVID-19-related morphological brain changes in individuals who recovered from mild-to-moderate COVID-19.
Collapse
Affiliation(s)
- Lukas Haider
- Section of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, University College London Institute of Neurology, London, UK
| | - Paulus Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Iscel Ahmet
- Fifth Medical Department with Cardiology, Favoriten Clinic, Vienna, Austria
| | - Alexandra Dena
- Section of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Alexander P Thurnher
- Section of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Emanuele Tomassino
- Section of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
- Department of Diagnostic Neuroradiology, Hôpital Neurologique - Université Claude Bernard 1, Lyon, France
| | - Diana Bonderman
- Fifth Medical Department with Cardiology, Favoriten Clinic, Vienna, Austria
- Department of Internal Medicine, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | - Stefan Seidel
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Majda Thurnher
- Section of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
21
|
Cho J, Park J, Son Y, Kim S, Jo H, Kyung S, Lee H, Yon DK. Post-Acute Sequelae of COVID-19 on Alopecia Areata in Individuals With Mental Illness in South Korea and Japan: A Binational Population-Based Cohort Study. J Med Virol 2025; 97:e70364. [PMID: 40275542 DOI: 10.1002/jmv.70364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/17/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
While previous studies have primarily focused on post-acute sequelae of COVID-19, the long-term effects of SARS-CoV-2 infection on alopecia areata (AA) among individuals with mental illness remain underexplored. Thus, this study aimed to address this gap by examining the association between post-acute sequelae of COVID-19 and AA, with a specific focus on individuals with mental illness. This study utilized bi-national, large-scale, and population-based cohorts of individuals with pre-existing mental illness: a Korean nationwide cohort (K-COV-N cohort; main cohort; total n = 3 248 448) and a Japanese claims-based cohort (JMDC cohort; replication cohort; total n = 696 332). The outcome focused on the new onset of AA following 30 days after SARS-CoV-2 infection, defined using ICD-10 codes L63. Using a propensity score-based overlap weighted algorithm, the adjusted hazard ratio (aHR) for AA following COVID-19 was calculated for individuals with mental illness. In the main cohort, the risk of AA as post-acute sequelae of COVID-19 was identified among individuals with mental illness (aHR, 1.32 [95% CI, 1.23-1.43]). The risk was significant for mild mental illness (aHR, 1.33 [95% CI, 1.24-1.44]) and within 6 months postinfection (aHR, 1.48 [95% CI, 1.35-1.63]). Similar findings were observed in the replication cohort. In conclusion, among individuals with mental illness, the risk of post-acute sequelae of COVID-19 on AA was elevated-particularly in those with mild mental illness-though this risk decreased over time. Our findings highlight the importance of early screening, integrated care, and equitable healthcare access for managing post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Jaehyeong Cho
- Department of Medicine, CHA University College of Medicine, Seongnam, South Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Jaeyu Park
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
- Department of Precision Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Yejun Son
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Precision Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Soeun Kim
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Precision Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Hyesu Jo
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
| | - Seoyeon Kyung
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
| | - Hayeon Lee
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Electronics and Information Convergence Engineering, Kyung Hee University, Yongin, South Korea
| | - Dong Keon Yon
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, South Korea
- Department of Regulatory Science, Kyung Hee University, Seoul, South Korea
- Department of Precision Medicine, Kyung Hee University School of Medicine, Seoul, South Korea
- Department of Electronics and Information Convergence Engineering, Kyung Hee University, Yongin, South Korea
- Department of Pediatrics, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, South Korea
| |
Collapse
|
22
|
Yang L, Zhou X, Liu J, Yang G, Yu J, Tan W, Fang X, Li W, He J, Ma Q, Yu L, Lu Z. Liang-Ge-San attenuates virus-induced acute lung injury by targeting FXR-mediated ACE2 downregulation to modulate the formation of the cytokine storm. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156584. [PMID: 40056637 DOI: 10.1016/j.phymed.2025.156584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Traditional Chinese medicine has been recognized for its significant role in treating acute lung injury (ALI) due to its distinct therapeutic advantages. Liang-Ge-San (LGS), a formulation from the ancient "Taiping Huimin Hejiju Fang", is believed to possess beneficial effects for treating ALI. However, LGS's precise mechanisms and efficacy in addressing viral ALI remain inadequately explored. PURPOSE To evaluate LGS's therapeutic effects and underlying mechanisms in treating viral-induced ALI. METHODS The protective effects of LGS were examined in a Polyinosinic-polycytidylic acid [Poly(I:C)]-induced ALI model using real-time quantitative PCR, enzyme-linked immunosorbent assay, and histopathological analysis. A bioinformatics approach combined with network pharmacology was utilized to ascertain the key targets of LGS in viral pneumonia. The pharmacodynamic mechanisms of LGS in viral ALI were further validated through immunofluorescence, overexpression, short hairpin RNA, chromatin immunoprecipitation, and target agonist assays. RESULTS LGS administration resulted in a reduction of IL-1β, IL-6, and TNF-α levels, along with a decrease in macrophage infiltration, pulmonary damage, and pneumonedema following the Poly(I:C) challenge. Bioinformatics and network pharmacology analyses suggested that Farnesyl X receptor (FXR) and angiotensin converting enzyme 2 (ACE2) are potential therapeutic targets for LGS in viral pneumonia. Further experiments revealed that LGS suppressed the expression of FXR, ACE2, and NF-κB-p65 in Poly(I:C)-infected cells. Notably, overexpression of FXR counteracted the repressive effects of LGS, while ACE2 expression remained unchanged in FXR-knockdown RAW264.7 cells upon treatment with Poly(I:C) or LGS. Additionally, LGS inhibited the interaction between FXR and ACE2 transcriptional promoters. In vivo, LGS attenuated the Poly(I:C)-induced upregulation of FXR, ACE2, IL-1β, IL-6, and TNF-α in ALI zebrafish and mice models, effects that could be reversed by chenodeoxycholic acid (CDCA), an FXR agonist. Moreover, LGS markedly alleviated weight loss, improved survival rates, reduced lung index, diminished viral load, and inhibited lung pathological changes in H1N1-PR8-induced ALI mice. IL-1β, IL-6, TNF-α, INF-γ, FXR, ACE2, small heterodimer partner, and NF-κB-p65 levels were markedly reduced by LGS, with these effects being reversed by CDCA. CONCLUSION This investigation provides the first evidence that FXR/ACE2 signaling is pivotal in acute respiratory viral infections, while LGS demonstrates antiviral activity against viral-induced ALI. LGS inhibits ACE2 expression induced by viral infection via FXR inhibition and modulates the cytokine storm, thus alleviating viral ALI. These findings suggest that LGS may be a promising treatment strategy for treating viral ALI.
Collapse
Affiliation(s)
- Liling Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Guangli Yang
- Department of Central Laboratory, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Weifu Tan
- Department of Neonatology, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Xiaochuan Fang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Wei Li
- Department of Neonatology, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Jiayang He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, PR China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, PR China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
23
|
Corona A, Simoncini S, Richini G, Gatti I, Santorsola C, Patroni A, Tomasini G, Capone A, Zendra E, Shuman M. Ig-M and Ig-A Enriched Ig-G Infusion as Adjuvant Therapy in the Critically ill Patients Experiencing SARS-CoV-2 Severe Infection. J Intensive Care Med 2025; 40:536-546. [PMID: 39648609 DOI: 10.1177/08850666241301689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Introduction: SARS-CoV-2 in patients who need Intensive Care (ICU) is associated with a mortality rate ranging from 10 to 40%-45%, with an increase in morbidity and mortality in presence of sepsis. Methods: We assumed that immunoglobulin (Ig) M and IgA enriched IgG (IGAM) therapy may support SARS COV-2 sepsis-related phase improving patient outcome. We conducted a retrospective case-control study on all the patients admitted to our ICU during the three pandemic waves between February 2020 and April 2021. Upon ICU admission, patients received anticoagulants with the standard supportive treatment (ST) ± IGAM therapy. After matching for the baseline characteristics and treatments, the patients receiving IGAM therapy too (group A), were compared with those undergoing ST (group B) only. Results: 85 patients were enrolled in group A, whereas 111 in group B. The mortality resulted lower in group A [37.6% versus 55.8%, OR: 0.7 (02-08), P = .01)]. A logistic regression analysis identified IGAM treatment as a survival predictor [OR: 0.35 (95%CI, 0.2-0.8)], whereas experiencing a super-infection [OR: 1.88 (95%CI, 1.5-4.9)] and a septic shock [OR: 1.92 (95%CI, 1.4-4.3)] as predictors of death. On day 7, the probability of dying was 3 times higher in patients treated with ST only. Variable life adjustment display (VLAD) was equal to 2.4 in group A, while - 2.2 group B (in terms of lives saved in relation with those expected, in according with Simplified Acute Physiology Score II (SAPS II) score. Conclusion: The treatment based on IGAM infusion seems to give an advantage chance of survival in SARS-CoV-2 severe infection. Further prospective studies are warranted.
Collapse
Affiliation(s)
- Alberto Corona
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Sara Simoncini
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Giuseppe Richini
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Ivan Gatti
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Clemente Santorsola
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Andrea Patroni
- Medical Directorate, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Giacomina Tomasini
- ICU, Anaesthesia and Emergency Department, ASST Valcamonica, Esine & Edolo Hospitals, Breno (BS), Italy
| | - Alice Capone
- ICU, Anaesthesia and Emergency Department, ASST Spedali Civili di Brescia, Brescia (BS), Italy
| | - Elena Zendra
- ICU, Anaesthesia and Emergency Department, ASST Spedali Civili di Brescia, Brescia (BS), Italy
| | - Myriam Shuman
- Department of Anaesthesiology, Pain Medicine and Perioperative Care, University of Washington, Seattle, Washington, USA
| |
Collapse
|
24
|
Zhu J, Li Y, Liu J, Zhou C, Liu X, Ding Y. Fatal Acute Necrotizing Encephalopathy in a 17-Year-Old Girl with COVID-19: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2025; 26:e946932. [PMID: 40308025 PMCID: PMC12054305 DOI: 10.12659/ajcr.946932] [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: 10/16/2024] [Accepted: 03/11/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Acute necrotizing encephalopathy (ANE) is a severe and rapidly progressive form of encephalopathy predominantly observed in children following various systemic infections. Although rare, cases of ANE have been increasingly associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This report details a fatal case of ANE in a 17-year-old girl diagnosed with coronavirus disease 2019 (COVID-19), emphasizing the critical need for early diagnosis and prompt intervention to improve clinical outcomes. CASE REPORT A previously healthy 17-year-old girl presented with a 3-day history of fever followed by onset of seizures and consciousness disorders after testing positive for SARS-CoV-2. Cranial magnetic resonance imaging (MRI) revealed bilateral symmetrical thalamus, which is a typical imaging presentation of ANE. Cerebrospinal fluid (CSF) examination revealed a normal cell count. Differential diagnoses such as acute disseminated encephalomyelitis (ADEM) and Leigh syndrome were excluded, leading to a diagnosis of ANE. Despite aggressive treatment, including respiratory support, intravenous immunoglobulin, and high-dose glucocorticoids, the patient's condition deteriorated rapidly, resulting in death the following day. CONCLUSIONS ANE is a rare but devastating condition in adolescents, particularly following viral infections such as COVID-19. It should be considered in patients presenting with seizures and progressive consciousness disorders after viral infection. Urgent neuroimaging and lumbar puncture are essential for diagnosis. Early recognition, immunotherapy, and aggressive management are crucial to mitigate the high mortality associated with this severe neurological disorder.
Collapse
Affiliation(s)
- Jianming Zhu
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, PR China
| | - Yanling Li
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, PR China
| | - Jianyi Liu
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, PR China
| | - Chaojun Zhou
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, PR China
| | - Xianglin Liu
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, PR China
| | - Yiyi Ding
- Department of Pediatrics, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, PR China
| |
Collapse
|
25
|
Keskitalo S, Seppänen MRJ, Del Sol A, Varjosalo M. From rare to more common: The emerging role of omics in improving understanding and treatment of severe inflammatory and hyperinflammatory conditions. J Allergy Clin Immunol 2025; 155:1435-1450. [PMID: 39978687 DOI: 10.1016/j.jaci.2025.02.011] [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: 10/07/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Inflammation is a pathogenic driver of many diseases, including atherosclerosis and rheumatoid arthritis. Hyperinflammation can be seen as any inflammatory response that is deleterious to the host, regardless of cause. In medicine, hyperinflammation is defined as severe, deleterious, and fluctuating systemic or local inflammation with presence of a cytokine storm. It has been associated with rare autoinflammatory disorders. However, advances in omics technologies, including genomics, proteomics, and metabolomics, have revealed it to be more common, occurring in sepsis and severe coronavirus disease 2019. With a focus on proteomics, this review highlights the key role of omics in this shift. Through an exploration of research, we present how omics technologies have contributed to improved diagnostics, prognostics, and targeted therapeutics in the field of hyperinflammation. We also discuss the integration of advanced technologies, multiomics approaches, and artificial intelligence in analyzing complex datasets to develop targeted therapies, and we address their potential for revolutionizing the clinical aspects of hyperinflammation. We emphasize personalized medicine approaches for effective treatments and outline challenges, including the need for standardized methodologies, robust bioinformatics tools, and ethical considerations regarding data privacy. This review aims to provide a comprehensive overview of the molecular mechanisms underpinning hyperinflammation and underscores the potential of omics technologies in enabling successful clinical management.
Collapse
Affiliation(s)
- Salla Keskitalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Mikko R J Seppänen
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland; Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; European Reference Network Rare Immunodeficiency Autoinflammatory and Autoimmune Diseases Network (ERN RITA) Core Center, Helsinki, The Netherlands
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Computational Biology Group, Basque Research and Technology Alliance (CIC bioGUNE-BRTA), Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland; Department of Biochemistry and Developmental Biology and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
26
|
Zhang L, Li B, Liu J, Bian YF, Lin GX, Zhou Y. Unveiling hub genes and biological pathways: A bioinformatics analysis of Trauma-Induced Coagulopathy (TIC). PLoS One 2025; 20:e0322043. [PMID: 40300035 PMCID: PMC12040245 DOI: 10.1371/journal.pone.0322043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Trauma-Induced Coagulopathy is a severe condition that rapidly manifests following traumatic injury and is characterized by shock, hypoperfusion, and vascular damage. This study employed bioinformatics methods to identify crucial hub genes and pathways associated with TIC. METHODS Microarray datasets (accession number GSE223245) were obtained from the Gene Expression Omnibus (GEO) database. The data were subjected analyses to identify the Differentially Expressed Genes (DEGs), which were further subjected to GO and KEGG pathway analyses. Subsequently, a Protein-Protein Interaction (PPI) network was constructed and hub DEGs closely linked to TIC were identified using CytoHubba, MCODE, and CTD scores. The diagnostic value of these hub genes was evaluated using Receiver Operating Characteristic (ROC) analysis. RESULTS Among the analyzed genes, 269 were identified as DEGs, comprising 103 upregulated and 739 downregulated genes. Notably, several significant hub genes were associated with the development of TIC, as revealed by bioinformatic analyses. CONCLUSIONS This study highlights the critical impact of newly discovered genes on the development and progression of TIC. Further validation through experimental research and clinical trials is required to confirm these findings.
Collapse
Affiliation(s)
- Lingang Zhang
- Emergency Department, Yuncheng Central Hospital affiliated to Shanxi Medical University,Yuncheng, Shanxi, China
| | - Bo Li
- Reproductive Medicine Department, Yuncheng Central Hospital affiliated to Shanxi Medical University, Yuncheng, Shanxi, China
| | - Jing Liu
- Pathology Department, Yuncheng Central Hospital affiliated to Shanxi Medical University,Yuncheng, Shanxi, China
| | - Yan feng Bian
- Emergency sungery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital,Third Hospital of Shanxi Medical University, China
| | - Guo xing Lin
- Emergency Department, Hebei province Xingtai Third People’s Hospital, Xingtai, China
| | - Ying Zhou
- Emergency Department, Yuncheng Central Hospital affiliated to Shanxi Medical University,Yuncheng, Shanxi, China
| |
Collapse
|
27
|
Souza HDS, Martins JSCC, Sousa TDC, Sardar S, Fintelman-Rodrigues N, Silva-Trujillo L, Souza TMLE, Siqueira MM, Fernandes JH, Matos ADR. Hypericin Suppresses SARS-CoV-2 Replication and Synergizes with Antivirals via Dual Targeting of RdRp and 3CLpro. Microorganisms 2025; 13:1004. [PMID: 40431177 PMCID: PMC12114490 DOI: 10.3390/microorganisms13051004] [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: 02/11/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
The continuous emergence of SARS-CoV-2 variants underscores the need for novel antiviral candidates. Hypericin (HY), a compound derived from Hypericum perforatum, exhibited potent in vitro activity against SARS-CoV-2 in Vero E6 cells, with low cytotoxicity (CC50 > 200 nM). HY showed no significant activity against Influenza A (H1N1) or dengue virus serotype 2, supporting its selective action. Antiviral effects were most evident when HY was administered post-infection, in a concentration-dependent manner, while cellular pretreatment or viral pre-incubation produced limited effects. Notably, HY also displayed virucidal activity, significantly reducing viral titers at 4 °C, 22 °C, and 37 °C. Combination treatments with remdesivir or nirmatrelvir enhanced antiviral efficacy by 50-70% relative to monotherapy, depending on compound concentration. Molecular simulations revealed stable interactions with conserved residues in RdRp and 3CLpro, suggesting a low risk of resistance. Together, these findings highlight the potential of HY as a selective antiviral and virucidal agent against SARS-CoV-2, particularly in combination regimens.
Collapse
Affiliation(s)
- Helena da Silva Souza
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Jéssica Santa Cruz Carvalho Martins
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Thiagos das Chagas Sousa
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Saiqa Sardar
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Natalia Fintelman-Rodrigues
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias, Oswaldo Cruz Institute, Rio de Janeiro 21040-361, Brazil; (N.F.-R.); (L.S.-T.); (T.M.L.e.S.)
- Center for Technological Development in Health, National Institute for Science and Technology on Innovation on Neglected Diseases Neglected Populations, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Lina Silva-Trujillo
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias, Oswaldo Cruz Institute, Rio de Janeiro 21040-361, Brazil; (N.F.-R.); (L.S.-T.); (T.M.L.e.S.)
- Center for Technological Development in Health, National Institute for Science and Technology on Innovation on Neglected Diseases Neglected Populations, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Thiago Moreno Lopes e Souza
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias, Oswaldo Cruz Institute, Rio de Janeiro 21040-361, Brazil; (N.F.-R.); (L.S.-T.); (T.M.L.e.S.)
- Center for Technological Development in Health, National Institute for Science and Technology on Innovation on Neglected Diseases Neglected Populations, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Marilda Mendonça Siqueira
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Jorge Hernandes Fernandes
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense (UENF), Campos dos Goytacazes 28013-602, Brazil
| | - Aline da Rocha Matos
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| |
Collapse
|
28
|
Modiga A, Butiurca VO, Boeriu CM, Truta TS, Turucz E, Halațiu VB, Rodean IP, Russu PC, Gherghinescu MC, Molnar C. Pathophysiological Mechanisms Linking COVID-19 and Acute Surgical Abdomen: A Literature Review. Life (Basel) 2025; 15:707. [PMID: 40430138 PMCID: PMC12113513 DOI: 10.3390/life15050707] [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: 03/18/2025] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Acute surgical abdomen is characterized by intense, sudden abdominal pain due to intra-abdominal conditions requiring prompt surgical intervention. The coronavirus disease 2019 (COVID-19) pandemic has led to various complications related to the disease's complex pathophysiological mechanisms, hence the hypothesis of COVID-19-induced acute abdominal surgical pathologies. The connection between acute surgical abdomen and COVID-19 involves two primary mechanisms. First, there is the presence of angiotensin-converting enzyme 2 (ACE2) receptors in multiple abdominal organs. This facilitates the cytokine storm through direct viral injury and inflammation. Second, the hypercoagulable state induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) increases the thrombotic risk within abdominal vessels, which can subsequently lead to ischemia. ACE2 receptors are notably expressed in the gastric, duodenal, and rectal epithelium, with SARS-CoV-2 viral RNA and nucleocapsid proteins detected in these tissues. The inflammatory response results in significant endothelial damage, activating coagulation pathways that cause monocellular infiltration, lymphocytic inflammation, and uncontrolled coagulation. These findings highlight the need for further research to clarify how COVID-19 leads to acute abdominal pathologies. Understanding these mechanisms is vital for improving clinical management and patient outcomes during future health crises and in the aftermath of the pandemic.
Collapse
Affiliation(s)
- Andrei Modiga
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Vlad-Olimpiu Butiurca
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Cristian Marius Boeriu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Teodora Sorana Truta
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Emilia Turucz
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Vasile-Bogdan Halațiu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
| | - Ioana-Patricia Rodean
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
| | - Paul Cristian Russu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Mircea Constantin Gherghinescu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Călin Molnar
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| |
Collapse
|
29
|
Xi Y, Zhou Z, Chang T, Dou G, Chu Z. Acute Macular Neuroretinopathy Mediated by COVID-19 Infection: Insights into its Clinical Features and Pathogenesis. FRONT BIOSCI-LANDMRK 2025; 30:26412. [PMID: 40302322 DOI: 10.31083/fbl26412] [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: 09/03/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 05/02/2025]
Abstract
Acute macular neuroretinopathy (AMN) is a rare retinal condition that predominantly affects young females. The incidence of AMN increased significantly during the COVID-19 pandemic, thereby providing a unique opportunity to elucidate the etiology of this disease. In the present study, 24 articles reporting 59 patients were reviewed. The average age of the patients was 33.51 ± 14.02 years, ranging from 16 to 75 years, with females comprising 71.19% of the cases. The average duration of ocular symptoms post-infection was 8.22 ± 10.69 days, ranging from 4 to 150 days. This study investigated the potential pathogenesis of AMN, including the impact of COVID-19 on retinal neurovascular structure and function, immune-mediated inflammatory factor production, blood-retinal barrier disruption, and retinal microvascular damage, as well as potential clinical therapeutic interventions. This research provides a theoretical framework that can inform further investigations of AMN.
Collapse
Affiliation(s)
- Yixuan Xi
- College of Life Sciences, Northwestern University, 710069 Xi'an, Shaanxi, China
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
- Department of Ophthalmology, First Affiliated Hospital of Northwest University, Xi'an First Hospital, 710002 Xi'an, Shaanxi, China
| | - Ziyi Zhou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Tianfang Chang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Guorui Dou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, 710032 Xi'an, Shaanxi, China
| | - Zhaojie Chu
- Department of Ophthalmology, First Affiliated Hospital of Northwest University, Xi'an First Hospital, 710002 Xi'an, Shaanxi, China
- Shaanxi Institute of Ophthalmology, 710021 Xi'an, Shaanxi, China
| |
Collapse
|
30
|
da Silva-Neto PV, de Carvalho JCS, Toro DM, Oliveira BTM, Cominal JG, Castro RC, Almeida MA, Prado CM, Arruda E, Frantz FG, Ramos AP, Ciancaglini P, Martins RB, da Silveira JC, Almeida F, Malmegrim KCR, Sorgi CA. TREM-1-Linked Inflammatory Cargo in SARS-CoV-2-Stimulated Macrophage Extracellular Vesicles Drives Cellular Senescence and Impairs Antibacterial Defense. Viruses 2025; 17:610. [PMID: 40431622 PMCID: PMC12115590 DOI: 10.3390/v17050610] [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: 02/05/2025] [Revised: 03/29/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has significantly affected global health, with severe inflammatory responses leading to tissue damage and persistent symptoms. Macrophage-derived extracellular vesicles (EVs) are involved in the modulation of immune responses, but their involvement in SARS-CoV-2-induced inflammation and senescence remains unclear. Triggering receptors expressed on myeloid cell-1 (TREM-1) are myeloid cell receptors that amplify inflammation, described as a biomarker of the severity and mortality of COVID-19. This study investigated the composition and effects of macrophage-derived EVs stimulated by SARS-CoV-2 (MφV-EVs) on the recipient cell response. Our results, for the first time, show that SARS-CoV-2 stimulation modifies the cargo profile of MφV-EVs, enriching them with TREM-1 and miRNA-155 association, along with MMP-9 and IL-8/CXCL8. These EVs carry senescence-associated secretory phenotype (SASP) components, promote cellular senescence, and compromise antibacterial defenses upon internalization. Our findings provide evidence that MφV-EVs are key drivers of inflammation and immune dysfunction, underscoring their potential as therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Pedro V. da Silva-Neto
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Jonatan C. S. de Carvalho
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Diana M. Toro
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (D.M.T.); (E.A.)
| | - Bianca T. M. Oliveira
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (B.T.M.O.); (F.A.)
| | - Juçara G. Cominal
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
| | - Ricardo C. Castro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Maria A. Almeida
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos-FZEA, Universidade de São Paulo-USP, Pirassununga 13635-900, SP, Brazil; (M.A.A.); (C.M.P.); (J.C.d.S.)
| | - Cibele M. Prado
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos-FZEA, Universidade de São Paulo-USP, Pirassununga 13635-900, SP, Brazil; (M.A.A.); (C.M.P.); (J.C.d.S.)
| | - Eurico Arruda
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (D.M.T.); (E.A.)
| | - Fabiani G. Frantz
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Ana P. Ramos
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (B.T.M.O.); (F.A.)
| | - Ronaldo B. Martins
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Juliano C. da Silveira
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos-FZEA, Universidade de São Paulo-USP, Pirassununga 13635-900, SP, Brazil; (M.A.A.); (C.M.P.); (J.C.d.S.)
| | - Fausto Almeida
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (B.T.M.O.); (F.A.)
| | - Kelen C. R. Malmegrim
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto-FCFRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-903, SP, Brazil; (R.C.C.); (F.G.F.); (R.B.M.); (K.C.R.M.)
| | - Carlos A. Sorgi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto 14040-901, SP, Brazil; (P.V.d.S.-N.); (J.C.S.d.C.); (J.G.C.); (A.P.R.); (P.C.)
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto-FMRP, Universidade de São Paulo-USP, Ribeirão Preto 14049-900, SP, Brazil; (B.T.M.O.); (F.A.)
- Programa de Pós-graduação em Imunologia Básica e Aplicada-PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas-UFAM, Manaus 69080-900, AM, Brazil
| |
Collapse
|
31
|
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
|
32
|
Chea M, de Langavant LC, Delorme C, Corvol JC, Delemazure J, Morawiec E, Faure M, Pichon B, Bayen E, Stefanescu F. Prevalence and typology of hallucinatory phenomena in adults with COVID-19: Early experiences at the post-acute phase in step down unit. Ann Phys Rehabil Med 2025; 68:101970. [PMID: 40252612 DOI: 10.1016/j.rehab.2025.101970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/01/2025] [Accepted: 02/08/2025] [Indexed: 04/21/2025]
Affiliation(s)
- Maryane Chea
- Department of Physical and Rehabilitation Medicine, Pitié-Salpêtrière Hospital, 75013 Paris, France; Groupe de Recherche Clinique GRC 24, Sorbonne Université, 75013 Paris, France
| | - Laurent Cleret de Langavant
- Global Brain Health Institute, Memory and Aging Center, University of California San Francisco, USA; Department of Neurology, Henri Mondor Hospital, 1, rue Gustave Eiffel, 94010 Créteil, France
| | - Cécile Delorme
- Department of Neurology, Pitié-Salpêtrière Hospital, 75013 Paris France
| | | | - Julie Delemazure
- Department of Pneumology, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Elise Morawiec
- Department of Pneumology, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Morgane Faure
- Department of Pneumology, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Bertrand Pichon
- Department of Physical and Rehabilitation Medicine, Pitié-Salpêtrière Hospital, 75013 Paris, France; Groupe de Recherche Clinique GRC 24, Sorbonne Université, 75013 Paris, France
| | - Eléonore Bayen
- Department of Physical and Rehabilitation Medicine, Pitié-Salpêtrière Hospital, 75013 Paris, France; Groupe de Recherche Clinique GRC 24, Sorbonne Université, 75013 Paris, France; Global Brain Health Institute, Memory and Aging Center, University of California San Francisco, USA.
| | - François Stefanescu
- Department of Physical and Rehabilitation Medicine, Pitié-Salpêtrière Hospital, 75013 Paris, France; Groupe de Recherche Clinique GRC 24, Sorbonne Université, 75013 Paris, France
| |
Collapse
|
33
|
Wu S, Kang M, Liu Y, Xiao Y, Deng J, Zhang W, Liao Q. Clinical and molecular characteristics of Staphylococcus aureus isolates from patients with COVID-19 in Southwest China. BMC Infect Dis 2025; 25:546. [PMID: 40247159 PMCID: PMC12004627 DOI: 10.1186/s12879-025-10868-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND This study investigated the clinical and molecular characteristics of Staphylococcus aureus (S. aureus) isolates from patients with coronavirus disease 19 (COVID- 19) at West China Hospital between December 1, 2022 and January 31, 2023. METHODS In total, 102 strains isolated from sputum, bronchoalveolar lavage fluid, endotracheal aspirates, and blood were collected from 102 patients and subjected to multilocus sequence typing and antimicrobial susceptibility testing. Eighteen virulence genes were also analyzed by polymerase chain reaction. RESULTS Seventy-five patients were discharged and 27 died. The predominant comorbidities were hypertension, diabetes mellitus, and cardiac disease. Twenty-eight known sequence types (STs) and 10 novel ones (ST8773/CC398, ST9221/CC5, ST9222/CC59, ST9223/CC8, ST9224/CC22, ST9225/CC1, ST9226/CC5, ST9227/CC59, ST9228/CC59, ST9229/CC398) were identified. The dominant molecular types were ST15 (CC15), ST59 (CC59), and ST5 (CC5). Among the three most prevalent STs, ST5 was significantly more resistant to levofloxacin, moxifloxacin, ciprofloxacin, and sulfamethoxazole than were ST59 and ST15. ST59 and ST5 had higher rates of resistance to erythromycin and clindamycin than ST15. All isolates contained at least eight virulence genes. The hemolysin gene hlb was found to be more prevalent in ST59 (100%) and ST5 (84.6%) than in ST15 (0) (P < 0.001). The prevalence of the enterotoxin gene seb in ST59 (100%) was significantly higher than that in ST5 (23.1%) and ST15 (20%) (P < 0.001), while the carrying rate of the sec gene was significantly higher in ST5 (76.9%) than that in ST59 (0) and ST15 (0) (P < 0.001). CONCLUSIONS The S. aureus isolates from patients with COVID- 19 in Southwest China exhibited a high degree of genetic diversity. Different STs exhibited different antimicrobial resistance patterns and virulence gene carrying rates.
Collapse
Affiliation(s)
- Siying Wu
- Division of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Mei Kang
- Division of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China.
| | - Ya Liu
- Division of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Yuling Xiao
- Division of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Jin Deng
- Division of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Weili Zhang
- Division of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Quanfeng Liao
- Division of Clinical Microbiology, Department of Laboratory Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| |
Collapse
|
34
|
Wu Y, Serna R, Gan W, Fan Z. Different patterns of leukocyte immune responses to infection of ancestral SARS-CoV-2 and its variants. Front Cell Infect Microbiol 2025; 15:1508120. [PMID: 40313462 PMCID: PMC12043629 DOI: 10.3389/fcimb.2025.1508120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
Background Contributions of leukocytes to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) defense have been reported extensively. However, it remains unclear whether there are different leukocyte responses to ancestral SARS-CoV-2 and its variants. Methods We analyzed peripheral blood leukocyte and subtype concentrations from 575 COVID-19 patients and 950 non-COVID-19 subjects registered at the University of Connecticut John Dempsey Hospital between 2020 and 2022, which covers the ancestral strain, Delta, and Omicron variants. Results We found that neutrophils, immature granulocytes, and monocytes were elevated, and lymphocytes were reduced after infection. These hyperactive neutrophils/immature granulocytes and suppressed lymphocytes/monocytes were associated with poorer prognosis in ancestral strain infection. Different from the ancestral strain, hyperactive immature granulocytes were not shown in the decedents of Delta infection, and immature granulocyte concentration was not observed to be associated with mortality. In Omicron infection, suppressed lymphocytes and monocytes were not shown in the decedents, and lymphocyte/monocyte concentrations were not associated with mortality. Conclusions Our findings provided insights into different leukocyte immune responses to ancestral SARS-CoV-2, Delta, and Omicron variants.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Raphael Serna
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Wenqi Gan
- Department of Public Health Sciences, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Zhichao Fan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
35
|
Kılıç Ö, Tecer D, Kaya MN, Çınar M, Yılmaz S. Can the Early Warning Score (ANDC) Predict Tocilizumab Efficacy in Patients with COVID-19 Cytokine Storm? Eur J Rheumatol 2025; 12:1-6. [PMID: 40377410 PMCID: PMC12060182 DOI: 10.5152/eurjrheum.2025.24048] [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: 05/30/2024] [Accepted: 11/22/2024] [Indexed: 05/18/2025] Open
Abstract
Background The aim of this study is to assess the effectiveness of the Early Warning Score [ANDC (age (A), neutrophil-to-lymphocyte ratio (NLR (N)), D-dimer (D), and CRP (C)] in predicting the treatment response in patients receiving tocilizumab for Coronavirus Disease 2019 (COVID-19)-related cytokine storm. Methods A retrospective review of medical records was conducted for patients treated with tocili- zumab for a cytokine storm related to COVID-19 between April 1, 2020, and April 1, 2021. Patient demographics, clinical characteristics, and laboratory parameters within 24 hours before tocilizumab were recorded. 1.14 × (age - 20) (years) + 1.63 × NLR + 5.00 × D-dimer (mg/L) + 0.14 × C-reactive protein (CRP) (mg/L) was used as the formula for the ANDC score. The study population was divided into 2 groups: those who died within 28 days of receiving tocilizumab and those who recovered. A comparative analysis was conducted. Results Within 28 days of tocilizumab treatment, 59 (35.32%) patients died. In comparison with living patients, deceased patients exhibited considerably higher levels of interleukin (IL)-6, lactate dehydro- genase (LDH), ANDC score, and CRP (P < .05). Lactate dehydrogenase was an independent predictor of response to tocilizumab treatment (P < .001) in a multivariate logistic regression analysis. In patients who did not receive steroid therapy before tocilizumab treatment, the ANDC score had the highest area under the curve (AUC). The optimal cut-off value was determined to be 92.56, with a sensitivity of 91.67% and a specificity of 60.61% (P < .001). In patients receiving steroids before tocilizumab, LDH had the highest AUC. The optimal cut-off value was 484.5 U/L (P < .001). Conclusion Lactate dehydrogenase was identified as an independent predictor of response to tocili- zumab treatment. The ANDC score showed the highest AUC value in steroid-naïve patients before tocilizumab, whereas LDH showed the highest AUC value in patients receiving steroids before tocili- zumab. Both the ANDC score and LDH levels show potential as valuable tools to guide treatment decisions.
Collapse
Affiliation(s)
- Özlem Kılıç
- Department of Rheumatology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Duygu Tecer
- Department of Rheumatology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Mehmet Nur Kaya
- Department of Rheumatology, Hakkari State Hospital, Hakkari, Türkiye
| | - Muhammet Çınar
- Department of Rheumatology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Sedat Yılmaz
- Department of Rheumatology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| |
Collapse
|
36
|
Rudroff T. Convergent Mechanisms in Virus-Induced Cancers: A Perspective on Classical Viruses, SARS-CoV-2, and AI-Driven Solutions. Infect Dis Rep 2025; 17:33. [PMID: 40277961 PMCID: PMC12027309 DOI: 10.3390/idr17020033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
This perspective examines the potential oncogenic mechanisms of SARS-CoV-2 through comparative analysis with established cancer-causing viruses, integrating classical virological approaches with artificial intelligence (AI)-driven analysis. The paper explores four key themes: shared oncogenic mechanisms between classical viruses and SARS-CoV-2 (including cell cycle dysregulation, inflammatory signaling, immune evasion, and metabolic reprogramming); the application of AI in understanding viral oncogenesis; the integration of neuroimaging evidence; and future research directions. The author presents novel hypotheses regarding SARS-CoV-2's potential oncogenic mechanisms, supported by recent PET/FDG imaging studies showing persistent metabolic alterations. The manuscript emphasizes the transformative potential of combining traditional virological methods with advanced AI technologies for better understanding and preventing virus-induced cancers, while highlighting the importance of long-term monitoring of COVID-19 survivors for potential oncogenic developments.
Collapse
Affiliation(s)
- Thorsten Rudroff
- Turku PET Centre, University of Turku, Turku University Hospital, 20520 Turku, Finland
| |
Collapse
|
37
|
Zhang W, Jing X, Li B, Wu X. Clearance of Cell-Free DNA: A Novel Target for Therapeutic Utilization in Multiple Systemic Disorders. ACS Biomater Sci Eng 2025; 11:2069-2079. [PMID: 40178087 DOI: 10.1021/acsbiomaterials.5c00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Cell-free DNA (cfDNA) holds significant promise for diagnostic and therapeutic advancements in medicine. This review delineates the utility of cfDNA in diagnostics and its therapeutic potential through clearance mechanisms for an array of diseases. Damage-associated molecular patterns (DAMPs) are endogenous molecules released by host cells during stress, or injury. As a trigger for inflammatory responses via damage-associated molecular patterns (DAMPs), cfDNA's removal via nanotechnological approaches can attenuate inflammation and promote tissue repair. While the application of cfDNA clearance is particularly auspicious in cancer, sepsis, and inflammatory conditions, it is confronted with challenges including toxicity, specificity, and the rigors of clinical trial validation. Collectively, this review delineates novel therapeutic targets to inform the development of innovative treatment strategies.
Collapse
Affiliation(s)
- Wenjun Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi030001, China
| | - Xuan Jing
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi030001, China
| | - Bing Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi030001, China
| | - Xiuping Wu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi030001, China
| |
Collapse
|
38
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Effects of oxidative stress on viral infections: an overview. NPJ VIRUSES 2025; 3:27. [PMID: 40295852 PMCID: PMC11993764 DOI: 10.1038/s44298-025-00110-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
Viral infections can trigger increased reactive oxygen species (ROS) production and a reduced antioxidant response in the host, leading to redox stress, inflammation, apoptosis, and ultimately, cell and tissue damage, which contribute to disease development. A better understanding of how ROS contributes to viral pathogenesis is critical for the development of novel therapeutic interventions. In this review, we discuss the current knowledge on ROS production and its effects across various viral infections, including severe acute respiratory syndrome-coronavirus-2, influenza A virus, dengue virus, Zika virus, hepatitis B virus, hepatitis C virus, and human immunodeficiency virus infections, to improve future therapeutic and preventive strategies for these infections.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh.
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, 890-0065, Japan.
| |
Collapse
|
39
|
Barušić Z, Bodulić K, Zember S, Laškaj R, Čivljak R, Puljiz I, Kurolt IC, Šafranko ŽM, Krajinović LC, Karić PS, Markotić A. Prognostic Value of Biomarkers in COVID-19: Associations with Disease Severity, Viral Variants, and Comorbidities-A Retrospective Observational Single-Center Cohort Study. Life (Basel) 2025; 15:634. [PMID: 40283188 PMCID: PMC12028838 DOI: 10.3390/life15040634] [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: 03/20/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Coronavirus disease (COVID-19) exhibits a wide spectrum of clinical severity and has been associated with specific biomarkers linked to disease progression and outcomes. This retrospective study analyzed sera from 1222 adult COVID-19 patients hospitalized at the University Hospital for Infectious Diseases in Croatia. We examined the association between several laboratory biomarker levels measured at patient admission and disease severity, fatal outcomes, viral variants and clinical parameters. Deceased patients and surviving patients with severe COVID-19 exhibited significantly elevated levels of several biomarkers on admission, including hs-troponin T, N-terminal pro-brain natriuretic peptide, creatine kinase, C-reactive protein, procalcitonin, interleukin-6, lactate dehydrogenase, lactate, urea and creatinine. Random forest models identified lymphocyte percentage, D-dimers, and hs-troponin T as the most important biomarkers for fatal outcome prediction, achieving 84.1% accuracy. Patients infected with the Delta SARS-CoV-2 variant exhibited significantly higher levels of proinflammatory, cardiac and renal biomarkers. Vaccination correlated with reduced proinflammatory parameters and higher lymphocyte proportions. Hypertension, chronic renal disease and diabetes were associated with increased cardiac, renal and metabolic biomarker levels, respectively. These findings highlight the association of several laboratory biomarkers with COVID-19 severity, viral variants, vaccination status and comorbidities, potentially offering prognostic insights into COVID-19 outcomes.
Collapse
Affiliation(s)
- Zoran Barušić
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
| | - Kristian Bodulić
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
| | - Sanja Zember
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
| | - Renata Laškaj
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
| | - Rok Čivljak
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Ivan Puljiz
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Ivan-Christian Kurolt
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
- Faculty of Medicine, Catholic University of Croatia, 10 000 Zagreb, Croatia
| | - Željka Mačak Šafranko
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
| | - Lidija Cvetko Krajinović
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
| | - Petra Svoboda Karić
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
| | - Alemka Markotić
- University Hospital for Infectious Diseases “Dr. Fran Mihaljević”, 10 000 Zagreb, Croatia; (Z.B.); (K.B.); (S.Z.); (R.L.); (R.Č.); (I.P.); (I.-C.K.); (Ž.M.Š.); (L.C.K.); (P.S.K.)
- Faculty of Medicine, Catholic University of Croatia, 10 000 Zagreb, Croatia
- Faculty of Medicine, University of Rijeka, 51 000 Rijeka, Croatia
| |
Collapse
|
40
|
Martino C, Kellman BP, Sandoval DR, Clausen TM, Cooper R, Benjdia A, Soualmia F, Clark AE, Garretson AF, Marotz CA, Song SJ, Wandro S, Zaramela LS, Salido RA, Zhu Q, Armingol E, Vázquez-Baeza Y, McDonald D, Sorrentino JT, Taylor B, Belda-Ferre P, Das P, Ali F, Liang C, Zhang Y, Schifanella L, Covizzi A, Lai A, Riva A, Basting C, Broedlow CA, Havulinna AS, Jousilahti P, Estaki M, Kosciolek T, Kuplicki R, Victor TA, Paulus MP, Savage KE, Benbow JL, Spielfogel ES, Anderson CAM, Martinez ME, Lacey JV, Huang S, Haiminen N, Parida L, Kim HC, Gilbert JA, Sweeney DA, Allard SM, Swafford AD, Cheng S, Inouye M, Niiranen T, Jain M, Salomaa V, Zengler K, Klatt NR, Hasty J, Berteau O, Carlin AF, Esko JD, Lewis NE, Knight R. SARS-CoV-2 infectivity can be modulated through bacterial grooming of the glycocalyx. mBio 2025; 16:e0401524. [PMID: 39998226 PMCID: PMC11980591 DOI: 10.1128/mbio.04015-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
The gastrointestinal (GI) tract is a site of replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and GI symptoms are often reported by patients. SARS-CoV-2 cell entry depends upon heparan sulfate (HS) proteoglycans, which commensal bacteria that bathe the human mucosa are known to modify. To explore human gut HS-modifying bacterial abundances and how their presence may impact SARS-CoV-2 infection, we developed a task-based analysis of proteoglycan degradation on large-scale shotgun metagenomic data. We observed that gut bacteria with high predicted catabolic capacity for HS differ by age and sex, factors associated with coronavirus disease 2019 (COVID-19) severity, and directly by disease severity during/after infection, but do not vary between subjects with COVID-19 comorbidities or by diet. Gut commensal bacterial HS-modifying enzymes reduce spike protein binding and infection of authentic SARS-CoV-2, suggesting that bacterial grooming of the GI mucosa may impact viral susceptibility.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for coronavirus disease 2019, can infect the gastrointestinal (GI) tract, and individuals who exhibit GI symptoms often have more severe disease. The GI tract's glycocalyx, a component of the mucosa covering the large intestine, plays a key role in viral entry by binding SARS-CoV-2's spike protein via heparan sulfate (HS). Here, using metabolic task analysis of multiple large microbiome sequencing data sets of the human gut microbiome, we identify a key commensal human intestinal bacteria capable of grooming glycocalyx HS and modulating SARS-CoV-2 infectivity in vitro. Moreover, we engineered the common probiotic Escherichia coli Nissle 1917 (EcN) to effectively block SARS-CoV-2 binding and infection of human cell cultures. Understanding these microbial interactions could lead to better risk assessments and novel therapies targeting viral entry mechanisms.
Collapse
Affiliation(s)
- Cameron Martino
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| | - Benjamin P. Kellman
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, USA
| | - Daniel R. Sandoval
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA
| | - Thomas Mandel Clausen
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA
- Copenhagen Center for Glycomics, Department of Molecular and Cellular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert Cooper
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Alhosna Benjdia
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, 78350, Jouy-en-Josas, France
| | - Feryel Soualmia
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, 78350, Jouy-en-Josas, France
- Sorbonne Université, Faculty of Sciences and Engineering, IBPS, UMR 8263 CNRS-SU, ERL INSERM U1345, Development, Adaptation and Ageing, F-75252 Paris, France
| | - Alex E. Clark
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Aaron F. Garretson
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Clarisse A. Marotz
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Se Jin Song
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| | - Stephen Wandro
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| | - Livia S. Zaramela
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Department of Biochemistry, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rodolfo A. Salido
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Qiyun Zhu
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Erick Armingol
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, USA
| | - Yoshiki Vázquez-Baeza
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Jacobs School of Engineering, University of California San Diego, La Jolla, California, USA
| | - Daniel McDonald
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - James T. Sorrentino
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, USA
| | - Bryn Taylor
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, USA
| | - Pedro Belda-Ferre
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Promi Das
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Farhana Ali
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Chenguang Liang
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
- Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Yujie Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
- Department of Biological & Medical Informatics, University of California San Francisco, School of Pharmacy, San Francisco, California, USA
| | - Luca Schifanella
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Animal Models and Retroviral Vaccine Section, Bethesda, Maryland, USA
| | - Alice Covizzi
- Department of Infectious diseases, Luigi Sacco Hospital, Milan and Department of Biomedical and Clinical Sciences (DIBIC), University of Milan, Milan, Italy
| | - Alessia Lai
- Department of Infectious diseases, Luigi Sacco Hospital, Milan and Department of Biomedical and Clinical Sciences (DIBIC), University of Milan, Milan, Italy
| | - Agostino Riva
- Department of Infectious diseases, Luigi Sacco Hospital, Milan and Department of Biomedical and Clinical Sciences (DIBIC), University of Milan, Milan, Italy
| | - Christopher Basting
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Courtney Ann Broedlow
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Aki S. Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki and Turku, Finland
- Institute for Molecular Medicine Finland, FIMM - HiLIFE, Helsinki, Finland
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki and Turku, Finland
| | - Mehrbod Estaki
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Tomasz Kosciolek
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Sano Centre for Computational Medicine, Krakow, Poland
| | - Rayus Kuplicki
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
| | | | | | - Kristen E. Savage
- Division of Health Analytics, Department of Computational and Quantitative Medicine, City of Hope, Duarte, California, USA
| | - Jennifer L. Benbow
- Division of Health Analytics, Department of Computational and Quantitative Medicine, City of Hope, Duarte, California, USA
- UC Health Data Warehouse, University of California Irvine, Irvine, California, USA
| | - Emma S. Spielfogel
- Division of Health Analytics, Department of Computational and Quantitative Medicine, City of Hope, Duarte, California, USA
| | - Cheryl A. M. Anderson
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California, USA
| | - Maria Elena Martinez
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California, USA
| | - James V. Lacey
- Division of Health Analytics, Department of Computational and Quantitative Medicine, City of Hope, Duarte, California, USA
| | - Shi Huang
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Niina Haiminen
- IBM T. J. Watson Research Center, Yorktown Heights, New York, USA
| | - Laxmi Parida
- IBM T. J. Watson Research Center, Yorktown Heights, New York, USA
| | - Ho-Cheol Kim
- AI and Cognitive Software, IBM Research-Almaden, San Jose, California, USA
| | - Jack A. Gilbert
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Daniel A. Sweeney
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sarah M. Allard
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Austin D. Swafford
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- International Biomedical Research Alliance, Bethesda, Maryland, USA
| | - Susan Cheng
- Division of Cardiology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael Inouye
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki and Turku, Finland
- Division of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Mohit Jain
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki and Turku, Finland
| | - Karsten Zengler
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Nichole R. Klatt
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jeff Hasty
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
- Molecular Biology Section, Division of Biological Science, University of California San Diego, La Jolla, California, USA
| | - Olivier Berteau
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, ChemSyBio, 78350, Jouy-en-Josas, France
| | - Aaron F. Carlin
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA
- Glycobiology Research and Training Center, University of California San Diego, La Jolla, California, USA
| | - Nathan E. Lewis
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
- Jacobs School of Engineering, University of California San Diego, La Jolla, California, USA
- Center for Molecular Medicine, Complex Carbohydrate Research Center, and Dept of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Rob Knight
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
41
|
Hagbi-Levi S, Abraham M, Gamaev L, Mishaelian I, Hay O, Zorde-Khevalevsky E, Wald O, Wald H, Olam D, Weiss L, Peled A. Identification of Dinaciclib and Ganetespib as anti-inflammatory drugs using a novel HTP screening assay that targets IFNγ-dependent PD-L1. Front Immunol 2025; 16:1502094. [PMID: 40264756 PMCID: PMC12011776 DOI: 10.3389/fimmu.2025.1502094] [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: 09/26/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction IFNγ plays both positive and negative roles in the regulation of innate and adaptive immune responses against tumors and virally infected tissues by upregulating CXCL10 and PD-L1 expression. Methods To identify novel pathways and drugs that regulate the IFNγ-dependent PD-L1, we expressed GFP under the control of mouse PD-L1 promoter in mouse cancer cells that up regulate PD-L1 and CXCL10 in response to IFNγ stimulation. Using these cells, we screened an FDA approved library of 1496 small molecules known for their ability to inhibit IFNγ-dependent increase in PD-L1. Results We identified 46 drugs that up regulated and 4 that down regulated IFNγ-dependent PD-L1 expression. We discovered that in addition to the known JAK inhibitors Ruxolitinib and Baricitinib, Dinaciclib, a CDK1/2/5/9 inhibitor, and Ganetespib, a Hsp90 inhibitor, significantly inhibit both PD-L1 and CXCL10 expression in the model cells. Furthermore, both drugs suppressed IFNγ-dependent CXCL10 and PD-L1 expression in-vitro in primary human lung cells and human cancer cells. These drugs also significantly inhibited delayed-type hypersensitivity (DTH) in-vivo in an inflammation mouse model. Discussion Our novel screening platform can therefore be used in the future to identify novel immunomodulators and pathways in cancer and inflammation, expanding therapeutic horizons.
Collapse
Affiliation(s)
- Shira Hagbi-Levi
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Lika Gamaev
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Inbal Mishaelian
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ophir Hay
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Elina Zorde-Khevalevsky
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ori Wald
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Hanna Wald
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Devorah Olam
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lola Weiss
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
42
|
Murakami T, Yamaguchi Y, Amiya S, Yoshimine Y, Nameki S, Okita Y, Kato Y, Hirata H, Takeda Y, Kumanogoh A, Morita T. CD147-high classical monocytes: a cellular biomarker for COVID-19 disease severity and treatment response. Inflamm Regen 2025; 45:8. [PMID: 40189583 PMCID: PMC11974131 DOI: 10.1186/s41232-025-00371-8] [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: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can lead to severe coronavirus disease 2019 (COVID-19), which is characterized by cytokine storm and organ dysfunction. The spike S1 subunit induces inflammatory cytokine production, but the immune cell subsets that respond to S1 stimulation and contribute to disease severity remain unclear. METHODS We analyzed serum samples and peripheral blood mononuclear cells (PBMCs) from patients with COVID-19 (moderate: n = 7; severe: n = 25) and healthy controls (n = 38). Using mass cytometry (cytometry by time-of-flight; CyTOF), we analyzed immune cell responses to S1 subunit stimulation in PBMCs from healthy donors and patients with COVID-19. We examined correlations among identified cell populations, serum cytokine levels, and clinical parameters. RESULTS Serum S1 subunit levels correlated with disease severity and inflammatory cytokine concentrations. S1 subunit stimulation induced dose-dependent cytokine production from PBMCs, predominantly from myeloid cells. CyTOF analysis identified classical monocytes with high CD147 expression (CD147hi cMono) as the primary source of S1-induced cytokines. The proportion of CD147hi cMono increased significantly in severe COVID-19 and decreased with clinical improvement. The frequency of CD147hi cMono showed a stronger positive correlation with clinical severity markers in younger patients compared to older patients. CONCLUSIONS CD147hi cMono are the primary cellular source of S1-induced inflammatory cytokines and may serve as potential biomarkers for monitoring COVID-19 severity and treatment response.
Collapse
Affiliation(s)
- Teruaki Murakami
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Yuta Yamaguchi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Saori Amiya
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Yuko Yoshimine
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Shinichiro Nameki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Yasutaka Okita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Medical Center for Translational Research, Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, 565-0871, Japan
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan.
- Strategic Global Partnership & X(Cross)-Innovation Initiative, Graduate School of Medicine, Osaka University and Osaka University Hospital, Osaka, Japan.
| |
Collapse
|
43
|
Zhang C, Luo H, Deng Y, Li H, Yu X, Liu J, Huang L, Yang X, Jiang Q. The clinical risk and post-COVID-19 sequelae in patients with myasthenia gravis: a retrospective observational study. Front Neurol 2025; 16:1513649. [PMID: 40264651 PMCID: PMC12012726 DOI: 10.3389/fneur.2025.1513649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Background There are indeed several studies addressing the severity of Coronavirus disease 2019 (COVID-19) infection in myasthenia gravis (MG) patients. However, data on post-COVID-19 sequelae in MG patients remain limited. To address this gap, we collected clinical data on the condition and prognosis of MG patients with COVID-19 infection, aiming to investigate factors influencing both the severity of the infection and the occurrence of post-COVID-19 sequelae at 1 and 12 months after recovery. Method This was a retrospective analysis of 150 MG patients with COVID-19 infection from November 1, 2022 to March 1, 2023 at the First Affiliated Hospital of Guangzhou University of Chinese Medicine, including patient demographics, clinical characteristics, and post-COVID-19 sequelae. Multivariable binary logistic and linear regression models were employed to ascertain the variables influencing the severity. The evolution of the post-COVID-19 sequelae was analyzed using McNemar's test. Result In 150 MG patients, 128 (85.3%) patients were presented with COVID-19 infection, and 23 (18%) patients were hospitalized. The severity was associated with the daily corticosteroid dose (OR = 1.08, p = 0.02) and the frequency of myasthenia crises pre-COVID-19 (b = 7.8, t = 2.14, p = 0.035). Compared to normal patients, MG patients are more likely to experience post-COVID-19 sequelae such as insomnia, myalgia, dizziness, cough, expectoration, and sore throat at 12 months after recovery. Among these, the prevalence of myalgia, dizziness, rash, and vision impairment was significantly higher. Conclusion Compared to normal patients, MG patients are prone to developing severe COVID-19 infection, which is associated with the daily corticosteroid dose and the frequency of myasthenia crises pre-COVID-19. Additionally, they are prone to experiencing post-COVID-19 sequelae, including insomnia, myalgia, dizziness, cough, expectoration, and sore throat, at 12 months after recovery.
Collapse
Affiliation(s)
- Chaoyue Zhang
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haocheng Luo
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yufei Deng
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongjin Li
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Yu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Liu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linqi Huang
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojun Yang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qilong Jiang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
44
|
Barrachina-Esteve O, Anguita A, Reverter A, Espinosa J, Lafuente C, Rubio-Roy M, Crosas M, Vila-Sala C, Acero C, Navarro M, Cánovas D, Ribera G, Jodar M, Estela J. Neurologic features in hospitalized patients with COVID-19: a prospective cohort in a catalan hospital. Neurol Sci 2025; 46:1477-1488. [PMID: 39951175 PMCID: PMC11920300 DOI: 10.1007/s10072-025-08031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVES To study the prevalence and timing of neurological manifestations, including cognitive involvement, in patients hospitalized for Coronavirus disease 2019 (COVID-19). To analyze the pathogenic mechanisms and any association they have with disease severity. METHODS Longitudinal cohort study with prospective follow-up of patients who required hospitalization. Patients under 65 who had no pre-existing cognitive impairment and did not require an ICU stay were evaluated 3 and 12 months after discharge using a battery of neuropsychological tests. RESULTS Of 205 patients hospitalized for COVID-19, 153 (74.6%) presented with neurological manifestations. The most frequent were myalgia (32.7%), headache (31.7%), dysgeusia (29.2%), and anosmia (24.9%). Patients with more severe illness at the time of hospitalization presented fewer neurological manifestations. Of the 62 patients who underwent neuropsychological examination 3 months after discharge, 22.6% had impaired attention, 19.4% impaired working memory, 16.1% impaired learning and retrieval, 9.7% impaired executive functions, and 8.2% impaired processing speed. Patients with anosmia also presented with more headache (OR 5.45; p < 0.001) and greater risk of working memory impairment (OR 5.87; p 0.03). At follow-up 12 months after hospital discharge, 14.3% of patients still showed impaired attention, 2.4% impaired working memory, 2.5% impaired executive functions, and 2.5% impaired processing speed. DISCUSSION Neurological manifestations are common in patients hospitalized for COVID-19 regardless of severity. The high prevalence of anosmia and its association with headache and working memory impairment at 3 months, suggest potential direct or indirect damage to the prefrontal cortex via invasion of the olfactory bulb by COVID-19.
Collapse
Affiliation(s)
- Oriol Barrachina-Esteve
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain.
- Department of Neurology, Manacor Hospital, Manacor, Mallorca, Spain.
| | - A Anguita
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
- Department of Neurology, Sant Joan de Déu Hospital, Althaia Foundation, Manresa, Spain
| | - A Reverter
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - J Espinosa
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Lafuente
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Rubio-Roy
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Crosas
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Vila-Sala
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Acero
- Department of Ophthalmology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Navarro
- Department of Infectious Diseases, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - D Cánovas
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - G Ribera
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Jodar
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
- Department of Clinical and Health Psychology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), National Institute of Health Carlos III, Madrid, Spain
| | - J Estela
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| |
Collapse
|
45
|
Soni DK, Cabrera-Luque J, Kar S, Ahmed A, Sen C, Devaney J, Biswas R. Suppression of miR-155 Attenuates Lung Cytokine Storm Induced by SARS-CoV-2 Infection. J Interferon Cytokine Res 2025; 45:150-161. [PMID: 39950973 DOI: 10.1089/jir.2024.0253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a deadly human viral disease with a high rate of infection, morbidity, and mortality. Although vaccines and antiviral treatments are available, hospitalizations remain steady, and concerns about long-term consequences persist. Therefore, there is a great urgency to develop novel therapies. Here, we analyzed the role of miR-155, one of the most powerful drivers of host antiviral responses including immune and inflammatory responses, in the pathogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Endogenous microRNAs (miRNAs, miRs) are key molecules in preventing viral entry and replication while building an antiviral cellular defense. Our study reveals that miR-155 expression is elevated in patients with COVID-19. Using a mouse model transgenic for human angiotensin-converting enzyme receptor 2, we evaluated the potential of anti-miR-155 therapy. Treating SARS-CoV-2-infected mice with anti-miR-155 significantly reduced miR-155 expression, improved survival, and slightly increased body weight. Notably, these mice showed altered expression of cytokines in the lungs. These findings suggest anti-miR-155 could be a promising therapy to mitigate the cytokine storm and long-lasting symptoms induced by SARS-CoV-2 infection, improving public health outcomes and enhancing global pandemic preparedness.
Collapse
Affiliation(s)
- Dharmendra Kumar Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | | | - Anwar Ahmed
- Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Chaitali Sen
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Islam MT, Mahjabin A, Islam MM, Tasnim A, Al-Mahmood MR, Khasru MR, Salek AKM, Uddin T. Assessment of Post-COVID-19 Functional Status and Complications Among Survivors at a Tertiary Healthcare Center in Bangladesh. Cureus 2025; 17:e82866. [PMID: 40432639 PMCID: PMC12107017 DOI: 10.7759/cureus.82866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Background and objectives COVID-19 has caused widespread multisystem impairment, leaving survivors with significant post-COVID complications. Survivors have faced ongoing difficulties as a result of post-COVID complications. This study aimed to assess the post-COVID functional status and complications among COVID-19 survivors within a tertiary healthcare center in Bangladesh. Methodology In this observational study, 244 patients were selected based on predefined inclusion and exclusion criteria from the post-COVID-19 follow-up clinic of the Department of Medicine at Bangabandhu Sheikh Mujib Medical University (BSMMU). Results COVID-19 functional status was assessed using the Post-COVID-19 Functional Status Scale (PCFS). Most of the participants belonged to grade 1, whereas the fewest belonged to grade 4. Fatigue (190, 77.9%), sleep disturbances (126, 51.6%), anxiety (102, 41.8%), and breathing difficulties (98, 40.2%) were the most prevalent complications. Sleep disturbances and breathing difficulties were notably associated with most grades of functional status. Sleep disturbances showed statistical significance with all grades except grade 4 (P ≤ 0.05), while breathing difficulties were significantly associated with all grades except grade 2 (P ≤ 0.05). Conclusions The results of this study shed light on the long-term consequences of COVID-19 in a Bangladeshi context and underscore the importance of continued care and support for survivors. Timely interventions and rehabilitation services can play a crucial role in improving the overall quality of life for COVID-19 survivors in Bangladesh and beyond.
Collapse
Affiliation(s)
- Mohammad Tariqul Islam
- Physical Medicine and Rehabilitation, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | | | - Md Mahbubul Islam
- Physical Medicine and Rehabilitation, Manikganj Sadar Hospital, Manikganj, BGD
| | - Anika Tasnim
- Public Health and Informatics, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Md Rashid Al-Mahmood
- Physical Medicine and Rehabilitation, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
- Physical Medicine and Rehabilitation, Bangladesh Medical College, Dhaka, BGD
- Physical Medicine and Rehabilitation, Northern International Medical College, Dhaka, BGD
| | - Moshiur R Khasru
- Physical Medicine and Rehabilitation, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - A K M Salek
- Physical Medicine and Rehabilitation, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Taslim Uddin
- Physical Medicine and Rehabilitation, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| |
Collapse
|
47
|
Jeong DC, Lee SY. Macrophage activation syndrome in Kawasaki disease: a literature review of Korean studies. JOURNAL OF RHEUMATIC DISEASES 2025; 32:105-112. [PMID: 40134551 PMCID: PMC11931280 DOI: 10.4078/jrd.2024.0118] [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: 10/17/2024] [Revised: 12/18/2024] [Accepted: 01/15/2025] [Indexed: 03/27/2025]
Abstract
Macrophage activation syndrome (MAS) is a rare but potentially life-threatening complication of Kawasaki disease (KD). In Korea, many studies on KD have been reported, but there are only a few studies on MAS complicating KD (MAS-KD). This study was conducted to provide the characteristics of MAS-KD patients in Korea through a literature review. A total of 23 Korean patients with MAS-KD from 10 papers were included in this study. All MAS-KD patients met the hemophagocytic lymphohistiocytosis (HLH)-2004 criteria and/or the 2016 MAS criteria. The incidence of MAS-KD in Korean children was 0.8%~1.1%, which is relatively low compared to North America (1.9%). MAS-KD patients had lower rates of KD-related features and higher rates of incomplete KD, coronary artery abnormalities, and intravenous immunoglobulin resistance than patients with KD without MAS. Notable laboratory abnormalities in MAS-KD include anemia, neutropenia, thrombocytopenia, hypoalbuminemia, increased hepatic transaminase levels, and hyperferritinemia. For treatment of MAS-KD, the HLH-2004 protocol (i.e., 40 weeks of complex chemotherapy) was applied to 15 patients (65%), which is a significantly greater than those treated with this protocol in other countries (35%). Two patients (9%) died during the HLH-2004 protocol. In actual practice, MAS may be underrecognized in patients with KD. Clinical suspicion is paramount for early diagnosis and timely treatment.
Collapse
Affiliation(s)
- Dae Chul Jeong
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- The Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Soo-Young Lee
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- The Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
48
|
Weinstein J, Jagan N, Lorthridge‐Jackson S, Hamer‐Maansson J, Squier P. Ruxolitinib for Emergency Treatment of COVID-19-Associated Cytokine Storm: Findings From an Expanded Access Study. THE CLINICAL RESPIRATORY JOURNAL 2025; 19:e70050. [PMID: 40197709 PMCID: PMC11976455 DOI: 10.1111/crj.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 01/02/2025] [Accepted: 01/18/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION This expanded access program (EAP) provided ruxolitinib (oral, selective Janus kinase [JAK]1/JAK2 inhibitor) for emergency treatment of COVID-19-associated cytokine storm in patients eligible for hospitalization (NCT04355793). METHODS Patients received ruxolitinib 5 mg twice daily (preferred regimen when tolerated) or once daily for ≤ 14 days, or until determination of no clinical benefit was made. Outcomes were clinical status, physician-assessed clinical benefit, and serious adverse event (SAE) incidence. RESULTS Of 312 patients, 45.5% achieved ≥ 1-point clinical status improvement. Physician-assessed clinical benefit was reported in 42.6% of evaluable patients. SAEs occurred in 42.9%, with 2.6% experiencing an SAE suspected to be ruxolitinib related. CONCLUSIONS Overall, some hospitalized patients with COVID-19-associated cytokine storm who received ruxolitinib experienced clinical status improvement; ruxolitinib was well tolerated. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT04355793.
Collapse
Affiliation(s)
- Jeffrey Weinstein
- Department of Clinical Quality and Infection Prevention and ControlKettering Health NetworkDaytonOhioUSA
| | - Nikhil Jagan
- Department of MedicineCreighton University School of MedicineOmahaNebraskaUSA
| | | | | | - Peg Squier
- Medical AffairsIncyte CorporationWilmingtonDelawareUSA
| |
Collapse
|
49
|
Lai C, Lu S, Yang Y, You X, Xu F, Deng X, Lan L, Guo Y, Kuang Z, Luo Y, Yuan L, Meng L, Wu X, Song Z, Jiang N. Myeloid-Driven Immune Suppression Subverts Neutralizing Antibodies and T Cell Immunity in Severe COVID-19. J Med Virol 2025; 97:e70335. [PMID: 40183283 PMCID: PMC11969634 DOI: 10.1002/jmv.70335] [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: 11/28/2024] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025]
Abstract
The objective of this study was to better understand immune failure mechanisms during severe acute respiratory syndrome coronavirus 2, SARS-CoV-2 infection, which are critical for developing targeted vaccines and effective treatments. We collected 34 cases representing different disease severities and performed high-quality single-cell TCR/BCR sequencing to analyze the peripheral immune cell profiles. Additionally, we assessed antibody-neutralizing activity through in vitro experiments. Our integrated multiomics analysis uncovers a profound immune paradox in severe COVID-19: hyperinflammation coexists with immunosuppression, driven by distinct yet interconnected dysregulatory mechanisms. Severe patients develop robust humoral immunity, evidenced by clonally expanded plasma cells producing neutralizing antibodies (e.g., IGHG1-dominated responses) and antigen-specific T cell activation. However, these protective responses are counteracted by myeloid-driven immunosuppression, particularly CD14+ HMGB2+ monocytes exhibiting metabolic reprogramming and HLA-DR downregulation, coupled with progressive T cell exhaustion characterized by IFN-γ/TNF-α hyperactivation and impaired antigen presentation. Importantly, prolonged viral persistence in severe cases arises from a failure to coordinate humoral and cellular immunity-antibody-mediated neutralization cannot compensate for defective cytotoxic T cell function and monocyte-mediated immune suppression. These findings highlight the necessity for therapeutic strategies that simultaneously enhance antibody effector functions (e.g., Fc optimization), restore exhausted T cells, and reverse myeloid suppression. They also highlight the importance of vaccines designed to elicit balanced B cell memory and durable T cell responses, which are critical to preventing severe disease progression. By addressing the dual challenges of hyperinflammation and immunosuppression, such approaches could restore immune coordination and improve outcomes in severe COVID-19.
Collapse
Grants
- This work was supported by the National Key Research and Development Program of China (2021YFC2501800, 2022YFA0806200, 2023YFC0872500, and 2024YFC3044600), the National Natural Science Foundation of China (82072214, 82272198, and 82202373), the Science and Technology of Shanghai Committee (21MC1930400, 22Y11900100, and 23Y31900100), and the Shanghai Municipal Health Commission (2023ZDFC0101).
Collapse
Affiliation(s)
- Cong Lai
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Su Lu
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yilin Yang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Xiaoyu You
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Feixiang Xu
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Xinran Deng
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Lulu Lan
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yuesheng Guo
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Zhongshu Kuang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yue Luo
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Li Yuan
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Lu Meng
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Xueling Wu
- Department of Respiratory MedicineShanghai Jiaotong University School of Medicine, Renji HospitalShanghaiChina
| | - Zhenju Song
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Ning Jiang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| |
Collapse
|
50
|
Nagoba BS, Dhotre SV, Gavkare AM, Mumbre SS, Dhotre PS. Convergence of COVID-19 and recurrent stroke: In-hospital mortality risks explored. World J Virol 2025; 14:99904. [PMID: 40134845 PMCID: PMC11612881 DOI: 10.5501/wjv.v14.i1.99904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
This editorial comments on the article by Desai et al, which investigates the impact of coronavirus disease 2019 (COVID-19) on in-hospital mortality among patients with recurrent stroke using data from the 2020 National Inpatient Sample. The findings reveal significantly higher mortality rates in COVID-19-positive patients compared to non-COVID-19 patients, particularly among middle-aged individuals, males, and ethnic minorities. This editorial explores the underlying mechanisms contributing to these outcomes and discusses the clinical implications for targeted management strategies in high-risk groups. The results emphasize the need for comprehensive approaches to mitigate the heightened risks faced by recurrent stroke patients during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Basavraj S Nagoba
- Department of Microbiology, Maharashtra Institute of Medical Sciences and Research (Medical College), Latur 413531, India
| | - Shree V Dhotre
- Department of Microbiology, Ashwini Rural Medical College, Solapur 413001, India
| | - Ajay M Gavkare
- Department of Physiology, Maharashtra Institute of Medical Sciences and Research (Medical College), Latur 413531, India
| | - Sachin S Mumbre
- Department of Community Medicine, Ashwini Rural Medical College, Solapur 413001, India
| | - Pradnya S Dhotre
- Department of Biochemistry, Ashwini Rural Medical College, Solapur 413001, India
| |
Collapse
|