1
|
Costa MVL, de Aguiar EJ, Rodrigues LS, Traina C, Traina AJM. DEELE-Rad: exploiting deep radiomics features in deep learning models using COVID-19 chest X-ray images. Health Inf Sci Syst 2025; 13:11. [PMID: 39741501 PMCID: PMC11683036 DOI: 10.1007/s13755-024-00330-6] [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: 01/15/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Purpose Deep learning-based radiomics techniques have the potential to aid specialists and physicians in performing decision-making in COVID-19 scenarios. Specifically, a Deep Learning (DL) ensemble model is employed to classify medical images when addressing the diagnosis during the classification tasks for COVID-19 using chest X-ray images. It also provides feasible and reliable visual explicability concerning the results to support decision-making. Methods Our DEELE-Rad approach integrates DL and Machine Learning (ML) techniques. We use deep learning models to extract deep radiomics features and evaluate its performance regarding end-to-end classifiers. We avoid successive radiomics approach steps by employing these models with transfer learning techniques from ImageNet, such as VGG16, ResNet50V2, and DenseNet201 architectures. We extract 100 and 500 deep radiomics features from each DL model. We also placed these features into well-established ML classifiers and applied automatic parameter tuning and a cross-validation strategy. Besides, we exploit insights into the decision-making behavior by applying a visual explanation method. Results Experimental evaluation on our proposed approach achieved 89.97% AUC when using 500 deep radiomics features from the DenseNet201 end-to-end classifier. Besides, our ensemble DEELE-Rad method improves the results up to 96.19% AUC for the 500 dimensions. To outperform, ML DEELE-Rad reached the best results with an Accuracy of 98.39% and 99.19% AUC for the same setup. Our visual assessment employs additional possibilities for specialists and physicians to decision-making. Conclusion The results reflect that the DEELE-Rad approach provides robustness and confidence to the images' analysis. Our approach can benefit healthcare specialists when employed at clinical routines and respective decision-making procedures. For reproducibility, our code is available at https://github.com/usmarcv/deele-rad.
Collapse
Affiliation(s)
- Márcus V. L. Costa
- Institute of Mathematics and Computer Science, University of São Paulo, São Carlos, São Paulo 13566-590 Brazil
| | - Erikson J. de Aguiar
- Institute of Mathematics and Computer Science, University of São Paulo, São Carlos, São Paulo 13566-590 Brazil
| | - Lucas S. Rodrigues
- Institute of Mathematics and Computer Science, University of São Paulo, São Carlos, São Paulo 13566-590 Brazil
| | - Caetano Traina
- Institute of Mathematics and Computer Science, University of São Paulo, São Carlos, São Paulo 13566-590 Brazil
| | - Agma J. M. Traina
- Institute of Mathematics and Computer Science, University of São Paulo, São Carlos, São Paulo 13566-590 Brazil
| |
Collapse
|
2
|
Jia S, Shao C, Cheng X, Pan H, Wang Z, Xia Y, Xu J, Huai X, Leng D, Wang J, Zhao G, Wang B, Li J, Zhu F. Immunogenicity and safety of a COVID-19 DNA vaccine in healthy adults and elderly: A randomized, observer-blind, placebo-controlled phase 2 trial. Hum Vaccin Immunother 2025; 21:2448405. [PMID: 39865693 PMCID: PMC11776483 DOI: 10.1080/21645515.2024.2448405] [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: 09/09/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
INO-4800 represents a DNA-based vaccine encoding the spike protein of SARS-CoV-2. This phase 2 trial evaluated the immunogenicity and safety of INO-4800 as a primary vaccination series in adults. We conducted a randomized, observer-blind, placebo-controlled phase 2 trial of intradermal injection of INO-4800 in both healthy adults and elderly individuals. Eligible participants from each age group were enrolled and randomly assigned in a 3:3:2 ratio to receive two doses of INO-4800 (1.0 mg or 2.0 mg) or placebo, followed by electroporation on day 0 and day 28. The primary immunogenicity endpoints focused on determining the geometric mean titers (GMTs) of spike-binding antibodies and live SARS-CoV-2 neutralizing antibody at day 30 after the second dose. The primary endpoint for safety was the occurrence of adverse events within 30 days after vaccination. A total of 781 volunteers were recruited and screened for eligibility, with 320 eligible young adults (≥18 to <60 years old) and 320 elderly (≥60 to ≤85 years old) were randomly assigned to receive the low-dose (1.0 mg, n = 120) or high-dose (2.0 mg, n = 120) INO-4800, or placebo (n = 80). Notably, both dose groups exhibited significant increases in spike-binding antibodies at day 30 after the second dose, with GMTs of 1609.3 (95% CI: 1385.5-1869.3) for the low-dose group and 3016.7 (95% CI: 2577.4-3530.8) for the high-dose group. Additionally, both dose groups induced neutralizing antibodies against live SARS-CoV-2, with GMTs of 4.7 (95% CI: 4.2-5.3) and 6.6 (95% CI: 5.9-7.4) at day 30 after the second dose. The incidence of adverse events within 30 days after vaccination was slightly higher in the high-dose group (115 [47.9%]) than that in the low-dose group (105 [43.8%]) (p = .0060). All adverse reactions were grade 1 or 2, primarily occurring within 14 days after vaccination. No vaccine-related serious adverse events were reported. The COVID-19 DNA vaccine INO-4800 at two doses (1.0 mg or 2.0 mg) showed an acceptable safety profile and modest immunogenicity, with the high-dose slightly more immunogenic than the low-dose.Clinical Trials Registration: www.chictr.org.cn, identifier is ChiCTR2000040146.
Collapse
MESH Headings
- Humans
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/adverse effects
- COVID-19 Vaccines/administration & dosage
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/adverse effects
- Male
- Adult
- Female
- Middle Aged
- Aged
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- SARS-CoV-2/immunology
- Young Adult
- Immunogenicity, Vaccine
- Spike Glycoprotein, Coronavirus/immunology
- Adolescent
- Aged, 80 and over
- Healthy Volunteers
- Vaccination
Collapse
Affiliation(s)
- Siyue Jia
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
| | - Chengwei Shao
- School of Public Health, Southeast University, Nanjing, China
| | - Xin Cheng
- R&D Business Unit, Advaccine Biopharmaceuticals Suzhou Co., Ltd, Suzhou, China
| | - Hongxing Pan
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
| | - Zhijian Wang
- Department of Acute Infectious Diseases and Immunization Program Management, Danyang Center for Disease Control and Prevention, Zhenjiang, China
| | - Yu Xia
- R&D Business Unit, Advaccine Biopharmaceuticals Suzhou Co., Ltd, Suzhou, China
| | - Jianfang Xu
- Department of Acute Infectious Diseases and Immunization Program Management, Danyang Center for Disease Control and Prevention, Zhenjiang, China
| | - Xuefen Huai
- R&D Business Unit, Advaccine Biopharmaceuticals Suzhou Co., Ltd, Suzhou, China
| | - Danjing Leng
- Department of Acute Infectious Diseases and Immunization Program Management, Danyang Center for Disease Control and Prevention, Zhenjiang, China
| | - Jiarong Wang
- R&D Business Unit, Advaccine Biopharmaceuticals Suzhou Co., Ltd, Suzhou, China
| | - Gan Zhao
- R&D Business Unit, Advaccine Biopharmaceuticals Suzhou Co., Ltd, Suzhou, China
| | - Bin Wang
- R&D Business Unit, Advaccine Biopharmaceuticals Suzhou Co., Ltd, Suzhou, China
| | - Jingxin Li
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
- School of Public Health, Southeast University, Nanjing, China
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, China
| | - Fengcai Zhu
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing, China
- School of Public Health, Southeast University, Nanjing, China
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Hsieh LC, Hsieh SL, Ping TN, Huang YC, Lin SJ, Chi HY, Wu CC. Apium graveolens L. alleviates acute lung injury in human A-549 cells by reducing NF-κB and NLRP3 inflammasome signaling. PHARMACEUTICAL BIOLOGY 2025; 63:1-13. [PMID: 39670672 DOI: 10.1080/13880209.2024.2433994] [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: 04/16/2024] [Revised: 10/10/2024] [Accepted: 11/20/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Apium graveolens L. (celery) is a dietary vegetable with anti-inflammatory properties. It has the potential to treat acute lung injury (ALI) caused by COVID-19 or other diseases. OBJECTIVE To investigate the effects of Apium graveolens water extract (AGWE) on ALI in human lung A-549 cells induced by lipopolysaccharide (LPS). MATERIALS AND METHODS A-549 cells were treated with AGWE for 24 h and then stimulated with 10 μg/mL LPS for another 24 h. The effects of AGWE on cell viability, the inflammatory response, oxidative stress, and apoptosis and their regulatory factors, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and NLR family pyrin domain containing 3 (NLRP3) inflammasome signaling activation were analyzed. RESULTS Treatment with 5-50 μg/mL AGWE reversed the decrease in cell viability caused by LPS (p < 0.05). AGWE can reduce interleukin (IL)-1β, IL-6, IL-18, and TNF-α levels; their EC50 values are 61.4, 65.7, 37.8, and 79.7 μg/mL, respectively. AGWE can reduce reactive oxygen species and thiobarbituric acid reactive substances in A-549 cells induced by LPS. AGWE also reduced the levels of apoptosis (EC50 of 74.8 μg/mL) and its regulators (Bid; Caspase-9, -8, and -3; Bax) and increased the levels of the mitochondrial membrane potential in A-549 cells induced by LPS. AGWE can also decrease the protein levels of NLRP3 and Caspase-1 and the activation of NF-κB signaling in A-549 cells induced by LPS. CONCLUSIONS These results show that 10 and 50 μg/mL AGWE can reduce the acute inflammation induced by LPS by reducing NF-κB and NLRP3 inflammasome signaling and mitochondria-dependent apoptosis pathways.
Collapse
Affiliation(s)
- Lan-Chi Hsieh
- Department of Dietetics, Kaohsiung Municipal United Hospital, Kaohsiung, Taiwan, R.O.C
| | - Shu-Ling Hsieh
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan, R.O.C
| | - Tsu-Ni Ping
- Department of Food and Nutrition, Providence University, Taichung, Taiwan, R.O.C
| | - Yi-Chun Huang
- Department of Food and Nutrition, Providence University, Taichung, Taiwan, R.O.C
| | - Ssu-Jung Lin
- Department of Food and Nutrition, Providence University, Taichung, Taiwan, R.O.C
| | - Hsing-Yu Chi
- Department of Food and Nutrition, Providence University, Taichung, Taiwan, R.O.C
| | - Chih-Chung Wu
- Department of Food and Nutrition, Providence University, Taichung, Taiwan, R.O.C
| |
Collapse
|
4
|
Ma L, Lin X, Xu M, Ke X, Liu D, Chen Q. Exploring the biological mechanisms of severe COVID-19 in the elderly: Insights from an aged mouse model. Virulence 2025; 16:2487671. [PMID: 40228062 PMCID: PMC12005417 DOI: 10.1080/21505594.2025.2487671] [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: 09/14/2024] [Revised: 02/04/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025] Open
Abstract
The elderly population, who have increased susceptibility to severe outcomes, have been particularly impacted by the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), leading to a global health crisis. However, definitive parameters or mechanisms underlying the severity of COVID-19 in elderly people remain unclear. Thus, this study seeks to elucidate the mechanism behind the increased vulnerability of elderly individuals to severe COVID-19. We employed an aged mouse model with a mouse-adapted SARS-CoV-2 strain to mimic the severe symptoms observed in elderly patients with COVID-19. Comprehensive analyses of the whole lung were performed using transcriptome and proteome sequencing, comparing data from aged and young mice. For transcriptome analysis, bulk RNA sequencing was conducted using an Illumina sequencing platform. Proteomic analysis was performed using mass spectrometry following protein extraction, digestion, and peptide labelling. We analysed the transcriptome and proteome profiles of young and aged mice and discovered that aged mice exhibited elevated baseline levels of inflammation and tissue damage repair. After SARS-CoV-2 infection, aged mice showed increased antiviral and inflammatory responses; however, these responses were weaker than those in young mice, with significant complement and coagulation cascade responses. In summary, our study demonstrates that the increased vulnerability of the elderly to severe COVID-19 may be attributed to an attenuated antiviral response and the overactivation of complement and coagulation cascades. Future research on antiviral and inflammatory responses is likely to yield treatments that reduce the severity of viral respiratory diseases in the elderly.
Collapse
Affiliation(s)
- Li Ma
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xian Lin
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Meng Xu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xianliang Ke
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Di Liu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Quanjiao Chen
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
5
|
Kaga M, Sasaki M, Masuda T, Sato H, Ueda T, Hirakawa A. Exploring the association between happy hypoxia and Coronavirus disease 2019 in the triage phase. Future Sci OA 2025; 11:2458413. [PMID: 39882841 PMCID: PMC11792821 DOI: 10.1080/20565623.2025.2458413] [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: 04/17/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Patients with severe coronavirus disease 2019 (COVID-19) have been reported to show hypoxia without displaying typical clinical signs or symptoms, called "happy hypoxia." To explore the potential of happy hypoxia as a distinctive symptom of COVID-19, we compared vital signs in the triage phase between patients with and without COVID-19. METHODS We retrospectively identified emergency patients with and without COVID-19 admitted to Rakuwakai Marutamachi Hospital, Kyoto, Japan, between January 2021 and December 2021. RESULTS AND CONCLUSIONS 317 patients were analyzed. Multivariate logistic regression analysis, including all vital signs, demonstrated that the respiratory rate was not statistically associated with COVID-19 (odds ratio, 0.94, p = 0.058), suggesting that happy hypoxia may not be a distinct hallmark of COVID-19.
Collapse
Affiliation(s)
- Mihiro Kaga
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
- Emergency and General Internal Medicine, Rakuwakai Marutamachi Hospital, Kyoto, Japan
| | - Masanao Sasaki
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Takahiro Masuda
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
- Department of Intensive Care, Institute of Science Tokyo, Tokyo, Japan
| | - Hiroyuki Sato
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Takeshi Ueda
- Emergency and General Internal Medicine, Rakuwakai Marutamachi Hospital, Kyoto, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Pini L, Giordani J, Levi G, Guerini M, Piva S, Peli E, Violini M, Piras S, El Masri Y, Pini A, Visca D, Assanelli D, Muiesan ML, Latronico N, Tantucci C, on behalf of the LOTO Investigators Working Group. Long-term alveolar-capillary diffusion impairments after severe SARS-CoV-2 pneumonia. Ann Med 2025; 57:2483383. [PMID: 40152750 PMCID: PMC11956098 DOI: 10.1080/07853890.2025.2483383] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Persistent respiratory symptoms and impaired gas exchange are common in patients recovering from COVID-19 pneumonia. The Lung Diffusing Capacity for Carbon Monoxide (DLCO) and Carbon Monoxide Transfer Coefficient (KCO) do not adequately distinguish alveolar membrane dysfunction from vascular abnormalities. This study aimed to characterize persistent diffusion impairment in post-ICU patients with prior SARS-CoV-2 pneumonia and reduced DLCO. METHODS After hospital discharge, patients underwent spirometry, DLCO measurement, and a 6-minute walking test every six months. If DLCO remained impaired at 18-24 months, a combined Lung Diffusing Capacity for Nitric Oxide (DLNO) and DLCO assessment was performed to differentiate alveolar-capillary membrane (DmCO) and pulmonary capillary blood volume (Vc) alterations. RESULTS Among 20 patients with persistent DLCO reduction, 3 had an obstructive ventilatory pattern, 6 had restriction, and 12 had low KCO. In restrictive cases, KCO was reduced but remained within normal limits without compensation. The DLNO/DLCO ratio exceeded 113.5% predicted in all patients. DmCO was impaired in 7 patients, while Vc was reduced in 16. CONCLUSION Both DLCO determinants were affected, with vascular impairment predominating. Vc reduction was present in most patients, with mean values below the lower limit of normality, whereas DmCO was less affected and often normal. The elevated DLNO/DLCO ratio suggests that persistent DLCO reduction is primarily driven by prolonged pulmonary capillary circulation dysfunction rather than alveolar membrane alterations, highlighting the vascular component as the primary site of long-term impairment.
Collapse
Affiliation(s)
- Laura Pini
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Respiratory Physiopathology Unit, ASST – Spedali Civili di Brescia, Brescia, Italy
| | - Jordan Giordani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Guido Levi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Pulmonology Department, ASST – Spedali Civili di Brescia, Brescia, Italy
| | - Michele Guerini
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Simone Piva
- Department of Anesthesia, Critical Care and Emergency, ASST Spedali Civili University Hospital, Brescia, Italy
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Elena Peli
- Department of Anesthesia, Critical Care and Emergency, ASST Spedali Civili University Hospital, Brescia, Italy
| | - Manuela Violini
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Piras
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Yehia El Masri
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Pini
- Department of Emergency, Anaesthesiological and Resuscitation Sciences, University Cattolica Sacro Cuore, Rome, Italy
| | - Dina Visca
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
- Department of Medicine and Cardiopulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Deodato Assanelli
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Internal Medicine Unit, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Maria Lorenza Muiesan
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Internal Medicine Unit, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Nicola Latronico
- Department of Anesthesia, Critical Care and Emergency, ASST Spedali Civili University Hospital, Brescia, Italy
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Claudio Tantucci
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | |
Collapse
|
7
|
Ferková S, Lepage M, Désilets A, Assouvie K, Lemieux G, Brochu I, Froehlich U, Gravel-Trudeau A, Vastra J, Jean F, Sarret P, Leduc R, Boudreault PL. Optimizing the pharmacokinetics and selectivity of TMPRSS2 inhibitors. Eur J Med Chem 2025; 294:117579. [PMID: 40382841 DOI: 10.1016/j.ejmech.2025.117579] [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: 02/18/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 05/20/2025]
Abstract
Since 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has undergone significant genomic mutations, contributing to resistance against existing 2019 coronavirus disease (COVID-19) treatments. In a previous study, we identified N-0385, a potent host-directed inhibitor of transmembrane serine protease 2 (TMPRSS2), which has therapeutic efficacy towards SARS-CoV-2 infection. However, in further evaluation of its preclinical druggability, N-0385 displayed unfavorable pharmacokinetic properties, including high bioavailability (99 %) following intranasal (IN) administration. This can lead to substantial systemic exposure and potential adverse effects due to off-target interactions. Here, we designed a library of peptidomimetic compounds with P3 site modifications on an optimized scaffold. We sought to maintain sub-nanomolar potency against TMPRSS2 (Kis < 2 nM), reduce pseudovirus infection, while addressing the lack of selectivity and excessive lung uptake. Notably, inhibitor 9, which contains Asp at the P3 position, achieved a two-fold increase in TMPRSS2 inhibitory potency (Ki = 0.13 ± 0.03 nM), a >700-fold selectivity over Factor Xa (FXa), and showed superior selectivity against other proteases (matriptase, transmembrane serine protease 6 (TMPRSS6), thrombin, furin, and tPA). Despite concerns about the role of FXa in the coagulation cascade, compound 9 had no impact on coagulation or thrombolysis 2 h after in vitro treatment. In the air-liquid interface (ALI) model of the lung epithelium, compound 9 displayed a 1.5-fold decrease in permeability compared to N-0385 and demonstrated sustained stability in lungs (11 h) and plasma (13 h). Taken together, our data demonstrate that continued optimization of this type of inhibitors will lead to improved therapeutics for the treatment of SARS-CoV-2 infection by IN administration.
Collapse
Affiliation(s)
- Sára Ferková
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Matthieu Lepage
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Antoine Désilets
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Kevin Assouvie
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gabriel Lemieux
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Isabelle Brochu
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Ulrike Froehlich
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Alice Gravel-Trudeau
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jules Vastra
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - François Jean
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
8
|
Kalbaugh DV. Dynamics of an epidemic controlled by isolation and quarantine: A probability-based deterministic model. Infect Dis Model 2025; 10:813-839. [PMID: 40271143 PMCID: PMC12017978 DOI: 10.1016/j.idm.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/25/2025] Open
Abstract
Assuming a homogeneous population, we employ a deterministic model based on first principles of probability to explore dynamics of an epidemic controlled by isolation alone, quarantine alone, and the two together. We develop explicit closed-form equations for key metrics of control performance: cumulative fraction of population infected over the course of the epidemic (final size), maximum fraction infected at any one time, and epidemic duration. We derive an analytical solution for final size of an epidemic controlled by isolation, when final size is small, and develop empirical relations for the other cases. We frame equations in terms of reproduction numbers, measures of intervention effort and initial conditions. We model both strength and speed of interventions, assume second order gamma distributions for intervention waiting times and employ non-time-invariant equations for quarantine. We also account for quarantine of unexposed, susceptible individuals and for imperfect intervention.
Collapse
|
9
|
Lan W, Tao L, Mao K, Song S, Liu H, Li Y, Wang X, Yu L, Xiong B. Porous poly (lactic-co-glycolic acid) microspheres loaded with neutral and acidic ginseng polysaccharides ameliorate pneumonia in mice. Carbohydr Polym 2025; 362:123697. [PMID: 40409812 DOI: 10.1016/j.carbpol.2025.123697] [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: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/30/2025] [Indexed: 05/25/2025]
Abstract
In this study, porous poly (lactic-co-glycolic acid) (PLGA) microspheres loaded with ginseng polysaccharides were successfully prepared and used to ameliorate pneumonia in mice. Fourier transform infrared spectroscopy, and confocal laser scanning microscopy confirmed the successful preparation of porous PLGA microspheres loaded with neutral ginseng polysaccharides (GP-N) and acidic ginseng polysaccharides (GP-S). Porous PLGA microspheres loaded with GP-N and GP-S had good release profiles. In vitro tests revealed that the release solution of porous PLGA microspheres loaded with GP-N and GP-S was nontoxic. The release solutions of porous PLGA microspheres loaded with GP-N and GP-S can both effectively reduce tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1β. Porous PLGA microspheres loaded with GP-N and GP-S ameliorated mouse pneumonia by modulating cytokine levels and antioxidant enzyme activities. Porous microspheres loaded with GP-S exhibited more potent anti-inflammatory effects than those loaded with GP-N. This phenomenon may be attributed to the presence of uronic acid in the GP-S. Transcriptomics revealed that the potential anti-pneumonia mechanism of porous PLGA microspheres loaded with GP-S may ameliorate pneumonia in mice through cytochrome P450 drug metabolism, arachidonic acid metabolism, and MAPK signaling pathways. These findings provide a theoretical basis for the amelioration of pneumonia using ginseng polysaccharides.
Collapse
Affiliation(s)
- Wenfei Lan
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Li Tao
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Kaixuan Mao
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Shixin Song
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - He Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Yanru Li
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Xiaotong Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Lei Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; National Engineering Research Center for Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, China.
| | - Boyu Xiong
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
10
|
Trier NH, Zivlaei N, Ostrowski SR, Sørensen E, Larsen M, Slibinskas R, Ciplys E, Frederiksen JL, Houen G. Virus-specific antibody responses in severe acute respiratory syndrome coronavirus 2-infected and vaccinated individuals. Immunol Lett 2025; 274:107004. [PMID: 40157431 DOI: 10.1016/j.imlet.2025.107004] [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: 08/05/2024] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can have a serious course with many complications, especially in immunocompromised individuals. In such persons, other latent virus infections may contribute to disease pathology, in particular viruses which infect immune cells such as Epstein-Barr virus (EBV) and cytomegalovirus (CMV). METHODS In this study, serology-based assays were conducted to analyse antibody responses to SARS-CoV-2 spike protein (SP), EBV Epstein-Barr nuclear antigen (EBNA)-1 and CMV phosphoprotein (pp)52 in naturally SARS-CoV-2-infected individuals, non-infected healthy controls (HCs) and vaccinated healthy controls (VHCs) to identify an association between SARS-CoV-2 antibodies and EBV and CMV antibodies in order to determine whether latent EBV and CMV infected individuals are more prone to become infected with SARS-CoV-2. Moreover, SARS-CoV-2, EBV, and CMV antibody responses were characterized in serum from patients with relapsing-remitting multiple sclerosis (RRMS), a chronic inflammatory disease strongly associated with EBV infections, to determine whether the serologic virus antibody profile varies in immunocompromised RRMS individuals upon SARS-CoV-2 vaccinations compared to VHCs. RESULTS Significantly elevated SP IgG, IgM and IgA levels were identified in SARS-CoV-2-infected immunocompetent individuals when compared to non-infected HCs. However, no correlation was found to serum antibodies between SARS-CoV-2, EBV, and CMV in individuals infected with SARS-CoV-2 and in VHCs, suggesting that latent infections with neither EBV nor CMV associates to SARS-CoV-2 infection. Moreover, no significant difference in SP IgG, IgA and IgM levels was observed between vaccinated RRMS patients and VHCs, indicating that the immune system of immune deficient RRMS patients and VHCs respond identical to SARS-CoV-2 vaccinations. CONCLUSION Collectively, SARS-CoV-2 SP antibody levels reflect the vaccination and infection history and do not associate with EBV and CMV serostatus.
Collapse
Affiliation(s)
- Nicole Hartwig Trier
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark.
| | - Nadia Zivlaei
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark.
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, BLegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark.
| | - Margit Larsen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark.
| | - Rimantas Slibinskas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Evaldas Ciplys
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Jette Lautrup Frederiksen
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, BLegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Gunnar Houen
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55 5230 Odense M, Denmark.
| |
Collapse
|
11
|
Ali N, Hasan RA, Ibrahim IA, Mahmoud MF. Agomelatine attenuates dexamethasone-induced neurotoxicity in rats through the activation of MT1/2 receptors and attenuation of oxidative stress. Eur J Pharmacol 2025; 998:177659. [PMID: 40274180 DOI: 10.1016/j.ejphar.2025.177659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/10/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
Previous studies showed that agomelatine ameliorates doxorubicin-induced brain injury in rats. Furthermore, it protects neurons against oxidative stress triggered by acute ischemia reperfusion injury. So, this study aimed to investigate the possible neuroprotective effects of agomelatine on dexamethasone-induced neurotoxicity in rats and the underlying mechanisms. Subcutaneous injections of dexamethasone (10 mg/kg, 4 days) were used to induce neurotoxicity in rats. Agomelatine (10 mg/kg), luzindole (2.5 mg/kg, a melatonin receptor blocker), and luzindole plus agomelatine treatment commenced 3 days before dexamethasone injections and concurrent with dexamethasone injections. Elevated plus maze test, Y-maze test and open field test were carried out after 1 h of the last dose of dexamethasone on day 7. On 8th day of the experiment, brain tissues were collected. Brain oxidative stress markers, immunohistochemical expression of β-amyloid and glial fibrillary acidic protein (GFAP) were measured. Moreover, histopathological changes in the cerebral cortex and hippocampus were recorded and the number of damaged cells was counted. Dexamethasone increased anxiety and memory impairment but decreased locomotor exploration activity. Furthermore, it increased brain oxidative stress, expression of β-amyloid and GFAP, increased the number of damaged neurons, and caused structural changes in cerebral cortex and hippocampus. All these deleterious changes were mitigated by agomelatine. Luzindole prior administration to agomelatine reversed the protective effects of agomelatine except its effect on lipid peroxidation. Collectively, these findings suggest that agomelatine can protect against dexamethasone-induced neurotoxicity partially by activating melatonin receptors in addition to exerting antioxidant effects.
Collapse
MESH Headings
- Animals
- Acetamides/pharmacology
- Acetamides/therapeutic use
- Dexamethasone/toxicity
- Oxidative Stress/drug effects
- Male
- Rats
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Receptor, Melatonin, MT2/metabolism
- Receptor, Melatonin, MT2/agonists
- Receptor, Melatonin, MT1/metabolism
- Receptor, Melatonin, MT1/agonists
- Neurotoxicity Syndromes/metabolism
- Neurotoxicity Syndromes/drug therapy
- Neurotoxicity Syndromes/etiology
- Neurotoxicity Syndromes/pathology
- Rats, Wistar
- Behavior, Animal/drug effects
- Glial Fibrillary Acidic Protein/metabolism
- Maze Learning/drug effects
- Naphthalenes
Collapse
Affiliation(s)
- Noura Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Rehab A Hasan
- Department of Histology, Faculty of Medicine for Girls, Al Azhar University, Cairo, 11751, Egypt
| | - Islam Ahmed Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Mona F Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
12
|
Hood O, Russell SL, Rahman M, Okwose NC, Jakovljevic DG, Roden LC. Respiratory function and sleep parameters in adults following recovery from acute COVID-19. Respir Med 2025; 243:108135. [PMID: 40319929 DOI: 10.1016/j.rmed.2025.108135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
The impact of COVID-19 on lung function and sleep in otherwise healthy individuals has been subject to a limited number of studies. The aim of this study was to investigate the effect of COVID-19 on pulmonary function and sleep in adults. Participants, 50-85 years old, who had recovered from COVID-19 (COVID-19 group: n = 48) and those without history of COVID-19 (control group: n = 28) underwent pulmonary function assessment (Forced Vital Capacity, FVC, and Slow Vital Capacity, SVC) using spirometry. Sleep and circadian variables were measured objectively with wrist-worn actigraphy for seven days. Subjective sleep of participants was assessed using the Pittsburgh Sleep Quality Index (PSQI). There were no significant differences in age (60 ± 6 vs 62 ± 6 years), BMI (26.30 ± 4.25 vs 26.48 ± 3.60 kg/m2), or pulmonary function (FVC, 4.02 ± 1.04 vs 3.80 ± 0.98 L, p = 0.36; and SVC, 3.82 ± 1.09 vs 3.89 ± 0.92 L, p = 0.76) between COVID-19 and control groups. The COVID-19 group had significantly reduced sleep efficiency (0.87 ± 0.04 vs 0.91 ± 0.04, p < 0.01), increased sleep disturbance (awakenings, 1.70 ± 1.02 vs 1.15 ± 1.15, p < 0.01; and wakefulness after sleep onset, 35:05 ± 25:37 vs 20:02 ± 12:48 min, p = 0.01) and PSQI score (5.19 ± 2.88 vs 3.93 ± 2.89, p = 0.01), compared to the control group. Individuals with history of COVID-19 demonstrate reduced sleep quality compared to a non-COVID-19 control group.
Collapse
Affiliation(s)
- Olivia Hood
- Clinical Sciences and Translational Medicine, Centre for Health and Life Sciences, Coventry University, Coventry, CV1 2DS, UK; School of Biosciences, College of Biomedical and Life Sciences, Cardiff University, UK
| | - Sophie L Russell
- Clinical Sciences and Translational Medicine, Centre for Health and Life Sciences, Coventry University, Coventry, CV1 2DS, UK; Department for Health, University of Bath, Bath, UK; Centre for Nutrition, Exercise and Metabolish, Univeristy of Bath, Bath, UK
| | - Mushidur Rahman
- Clinical Sciences and Translational Medicine, Centre for Health and Life Sciences, Coventry University, Coventry, CV1 2DS, UK
| | - Nduka C Okwose
- Clinical Sciences and Translational Medicine, Centre for Health and Life Sciences, Coventry University, Coventry, CV1 2DS, UK
| | - Djordje G Jakovljevic
- Clinical Sciences and Translational Medicine, Centre for Health and Life Sciences, Coventry University, Coventry, CV1 2DS, UK.
| | - Laura C Roden
- Clinical Sciences and Translational Medicine, Centre for Health and Life Sciences, Coventry University, Coventry, CV1 2DS, UK.
| |
Collapse
|
13
|
Yang G, Tan W, Yan L, Lao Q, Zheng W, Ding H, Yu J, Liu Y, Zou L, Guo M, Yu L, Zhou X, Li W, Yang L. Phillyrin for sepsis-related acute lung injury: A potential strategy suppressing GSK-3β. Mol Immunol 2025; 183:115-136. [PMID: 40359720 DOI: 10.1016/j.molimm.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
The efficacy of clinical drugs for acute lung injury/acute respiratory distress syndrome (ALI/ARDS) remains suboptimal. Phillyrin (PHN), a compound derived from Forsythia, is believed to alleviate sepsis-related ALI/ARDS; however, its mechanisms are not fully elucidated. In this study, we screened 8331 target genes associated with ALI/ARDS from public databases and identified six hub genes relevant to PHN treatment: AKT1, GSK-3β, PPP2CA, PPP2CB, PPP2R1A, and AR. Receiver operating characteristic analysis and single-cell sequencing analysis revealed the expression of AKT1, GSK-3β, PPP2CA, PPP2CB, and PPP2R1A were markedly elevated. Molecular docking and dynamics simulations indicated that PHN forms a structurally stable complex with glycogen synthase kinase-3β (GSK-3β). Mendelian randomization analyses suggested that PHN, as a potent GSK-3β inhibitor, may promote M2 macrophage polarization and reduce neutrophil recruitment. We validated these findings through in vivo and in vitro experiments, demonstrating that PHN lowers iNOS levels and raises MMR levels by downregulating GSK-3β mRNA expression and protein activity during lipopolysaccharide (LPS)-induced macrophage inflammation. Additionally, PHN inhibited GSK-3β mRNA expression and protein activity, reducing NF-κB-p65 nuclear translocation in LPS-induced zebrafish inflammation and mice ALI. This inhibition decreased levels of TNF-α and IL-6, increased IL-10 levels, promoted M2 macrophage polarization, suppressed neutrophil recruitment, and ultimately ameliorated ALI/ARDS. In conclusion, our results indicate that PHN effectively alleviates LPS-induced ALI/ARDS by suppressing GSK-3β signaling.
Collapse
Affiliation(s)
- 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, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Lijun Yan
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qiaocong Lao
- Central Laboratory, The Tenth Affiliated Hospital, Southern Medical University, Dongguan People's Hospital, Dongguan 523059, China
| | - Wujuan Zheng
- Department of Pharmacy, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Hongyan Ding
- Omega-3 Research and Conversion Center, Dongguan Innovation Research Institute, Guangdong Medical University, Dongguan 523900, China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yong Liu
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Liyi Zou
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Maorun Guo
- Pingyi Health Center of Pingyi County, Linyi 273300, China
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Wei Li
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China.
| | - Liling Yang
- Dongguan Municipal Key Laboratory for Precise Prevention and Treatment of Neonatal Severe Illnesses, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China; Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; Department of Pharmacy, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China.
| |
Collapse
|
14
|
Pontiroli AE, Ambrosio G, Leoni O, Forlani M, Antonelli B, Gronda E, Palazzuoli A, Bandera F, Galati G, Tagliabue E. Heart failure and co-morbidities confer a negative prognosis in COVID-19 infection. Int J Cardiol 2025:133492. [PMID: 40490033 DOI: 10.1016/j.ijcard.2025.133492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2025] [Revised: 06/01/2025] [Accepted: 06/06/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Since early reports, it has been shown that cardiovascular (CV) diseases, including heart failure (HF), represent a risk factor for infection, hospital admissions and mortality from COVID-19. The COVID-19 pandemics has been of major importance in Italy and in the Lombardy Region. Aims of this study were to compare COVID-19 infection in HF and No-HF subjects, and to quantify among HF patients the risk for COVID-19 infection and all-cause mortality. METHODS All consecutive patients (98,549) with at least one hospital discharge of HF (primary diagnosis) during January 1st, 2015, to December 31st, 2019, were identified in the Lombardy Region Database (>10 million inhabitants), and compared with No-HF subjects (394,104 with a lower age limit 40 years), randomly chosen in a 4:1 proportion among hospitalized patients. The whole cohort of cases of COVID-19 infection, laboratory-confirmed by RT-PCR, aged >40 years, diagnosed from the beginning of the epidemic on 21 February 2020 to 1 October 2020 was studied. The study outcomes were: occurrence, hospitalization, and death in COVID-19 cases. RESULTS Incidence of COVID-19 increased with age in both HF (p < 0.001) and No-HF patients (p < 0.001); cases (and incidence rates, IR) were 8648 (IR = 29.653 × 100.000) in HF and 14,256 (IR = 10.195) and in No-HF (p < 0.001); hospital admissions were 4974 (IR = 14.970) and 4943 (IR = 3.484), respectively (p 〈0001); deaths were 7650 (IR = 5.368) and 18,368 (IR = 56.921), respectively (p < 0.001); the incidence rate ratio (IRR) was 2.909 (95 % C.I. 2.908-2.909) for infection (p < 0.001), 4.297 (95 % C.I. 4.296-4.297) for hospital admission (p < 0.001), and 10.603 (95 % C.I.10.602-10.604) for mortality (p < 0.001). The excess IRR for mortality varied from 25.001 (95 % C.I. 24.971-25.032) for the age decade 40-49 to 1.925 (95 % C.I. 1.923-1.926) for the age decade 100-109. Among HF patients, age (OR = 1.087, 95 % C.I.1.05-1.088), male sex (OR = 1.27, 95 % C.I. 1.23-1.31), number of hospital admissions for HF during the period 2015-2019 (OR = 2.22, 95 % C.I. 2.11-2.33), co-morbidities (OR = 1.33, 95 % C.I. 1.32-1.35), or Charlson Index (OR = 1.21, 95 % C.I. 1.20-1.22), were risk factors for both infection and all-cause mortality at univariable and at multivariable analysis. CONCLUSION Infections, hospital admissions, and mortality for COVID-19 increased with age and male sex were more frequent in HF than in No-HF patients. Among HF patients, age and sex, number of hospital admissions for HF, co-morbidities, were risk factors for both infection and mortality. These data are of relevance for prioritizing interventions for prevention of infection, and for assistance to patients with COVID-19, and to inform management of future pandemics.
Collapse
Affiliation(s)
- Antonio E Pontiroli
- Università degli Studi di Milano, Dipartimento di Scienze della Salute, Milan, Italy.
| | - Giuseppe Ambrosio
- Università di Perugia, Dipartimento di Medicina - CERICLET, Istituto Nazionale Ricerche Cardiovascolari - INRC, Perugia, Italy; IRCCS MultiMedica, Milan, Italy
| | - Olivia Leoni
- Dipartimento della Salute, Regione Lombardia, Osservatorio Epidemiologico, Milan, Italy.
| | | | | | - Edoardo Gronda
- IRCCS Policlinico, U.O.C. Nefrologia, Dialisi e Trapianti di Rene, Milan, Italy
| | - Alberto Palazzuoli
- Unità Autonoma Malattie Cardiovascolari, Dipartimento Cardio-Toracico e Vascolare, Ospedale le Scotte, Universita di Siena, Siena, Italy.
| | - Francesco Bandera
- IRCCS MultiMedica, Milan, Italy; Università degli Studi di Milano, Dipartimento di Scienze Biomediche per la Salute, Milan, Italy.
| | | | | |
Collapse
|
15
|
Yan M, Zhan Q, Wu X, Zheng C, Liu D. Hepatic dysfunction in individuals with COVID-19 and its impact on pregnancy outcomes. Medicine (Baltimore) 2025; 104:e42745. [PMID: 40489829 DOI: 10.1097/md.0000000000042745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/11/2025] Open
Abstract
This study aimed to describe the incidence of abnormal liver function tests (LFTs) in pregnant women post-2019 coronavirus disease (COVID-19) and investigate the characteristics of pregnant women with abnormal LFTs and its impact on pregnancy outcomes. The data for 168 pregnant women who were infected with COVID-19 in the late stages of pregnancy were collected in Hefei Maternal and Child Health Hospital from December 2022 to January 2023, and 86 pregnant women with abnormal liver function were divided into the research group and 82 pregnant women with normal liver function into the control group for retrospective analysis. Population and laboratory data were collected and statistical analysis was conducted. Among the 168 pregnant women with COVID-19, 86 (51.2%) had elevated liver enzymes. In the control group, 4 (4.5%) had elevated liver enzymes. Differences between the 2 groups were statistically significant (P < .05). Single-factor analysis revealed that age, gestational week, and body mass index (BMI) exhibited statistically significant differences (P < .001) as potential factors influencing abnormal LFTs. Logistic regression analysis demonstrated that age (OR: 1.526), gestational week (OR: 1.321), and BMI (OR: 1.159) remained independent risk factors for liver injury (P < .05). Furthermore, the cesarean section rate, postpartum hemorrhage rate, rupture of membranes rate, and fetal intrauterine distress rate in the observation group were all significantly higher than those in the control group (P < .05). Additionally, the incidence of neonatal asphyxia, preterm birth, and low birth weight in the observation group were all significantly higher than those in the control group (P < .05). Pregnant individuals are at an elevated risk of hepatic injury following severe acute respiratory syndrome coronavirus 2 infection. Furthermore, the likelihood of hepatic injury escalates with advancing maternal age, gestational age, and BMI. Hepatic functional aberrations in the latter stages of pregnancy may precipitate adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Minqin Yan
- Department of Gynecology and Obstetrics, Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, P. R. China
| | | | | | | | | |
Collapse
|
16
|
Jánosi Á, Bódy B, Nagy R, Ocskay K, Kói T, Müller K, Túri I, Garami M, Hegyi P, Párniczky A. Tumour necrosis factor-alpha inhibitors decrease mortality in COVID-19: a systematic review and meta-analysis. Crit Care 2025; 29:232. [PMID: 40481519 PMCID: PMC12144840 DOI: 10.1186/s13054-025-05420-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/15/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Despite widespread vaccination efforts, effective treatment strategies remain critical for severe SARS-CoV-2 infection. Tumour necrosis factor-alpha (TNF-α) plays a central role in the cytokine storm characteristic of severe COVID-19. This systematic review and meta-analysis evaluates the effectiveness, efficacy, and safety of TNF-α inhibitors in the management of COVID-19. PATIENTS AND METHODS A systematic review of PubMed, Embase, and CENTRAL was conducted, focusing on studies involving SARS-CoV-2-infected patients treated with TNF-α inhibitors compared with those receiving standard of care without prior TNF-α inhibitor use. Data from studies published up to August 12, 2024, were analysed. Outcomes assessed included mortality, invasive mechanical ventilation, and C-reactive protein (CRP) levels. Odds ratios (ORs) and mean differences (MD) were calculated with 95% confidence intervals (CI), and subgroup analyses were performed for randomised controlled trials (RCTs) and non-randomised studies. RESULTS Seven studies involving 1393 patients with moderate-to-critical COVID-19 were included. TNF-α inhibitor treatment was associated with a reduced odds of mortality (OR 0.67, 95% CI [0.44-1.00], P = 0.052), which was statistically significant in the RCT subgroup across three studies (OR 0.75, 95% CI [0.58-0.97], P = 0.042, certainty of evidence: very low). The number needed to treat for mortality was calculated to be 16 (95% CI 9.0-inf.), which indicates that one additional death could be avoided for every 16 patients treated with TNF-α inhibitors compared to standard of care. No significant reduction in the need for invasive mechanical ventilation was observed (OR 0.95 [95% CI 0.46-1.94]; P = 0.822). Additionally, TNF-α inhibitors resulted in a significant reduction in CRP levels (MD - 21.9 mg/L [95% CI - 38.46 to - 5.34]; P = 0.024) within three to seven days post-treatment. CONCLUSION Our study indicates a potential role for TNF-α inhibition in the treatment of COVID-19 as their use was associated with reduced mortality, but further studies are needed to provide robust evidence.
Collapse
Affiliation(s)
- Ágoston Jánosi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Paediatric Institute, Budapest, Hungary
| | - Blanka Bódy
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Paediatric Institute, Budapest, Hungary
| | - Rita Nagy
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Paediatric Institute, Budapest, Hungary
| | - Klementina Ocskay
- Heim Pál National Paediatric Institute, Budapest, Hungary
- Pharmaceutical Sciences and Health Technologies Division, Doctoral School, Semmelweis University, Budapest, Hungary
| | - Tamás Kói
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Stochastics, Institute of Mathematics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Katalin Müller
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Paediatric Institute, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Department of Family Care Methodology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Ibolya Túri
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- András Pető Faculty, Semmelweis University, Budapest, Hungary
| | - Miklós Garami
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Paediatric Centre, Semmelweis University, Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Andrea Párniczky
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary.
- Heim Pál National Paediatric Institute, Budapest, Hungary.
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary.
| |
Collapse
|
17
|
Duong KE, Lu JY, Wang S, Duong TQ. Incidence and risk factors of new clinical disorders in patients with COVID-19 hyperinflammatory syndrome. Sci Rep 2025; 15:19892. [PMID: 40481056 PMCID: PMC12144132 DOI: 10.1038/s41598-025-04070-9] [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: 07/08/2024] [Accepted: 05/24/2025] [Indexed: 06/11/2025] Open
Abstract
This study investigated new incident clinical disorders in patients with COVID-19-related hyperinflammatory syndrome (cHIS) 3.5 years post infection. We analyzed 14,335 patients hospitalized with COVID-19 from March-2020 to July-2023. cHIS was defined based on a point system that included elevated body temperature, macrophage activation, hematological dysfunction, coagulopathy, and hepatic enzyme. Outcomes were newly diagnosed disorders of hypertension, diabetes, cardiovascular diseases, chronic kidney disease (CKD), chronic obstructive pulmonary disease (COPD), and asthma post COVID-19. Cumulative incidences and hazard ratios were computed. Compared to non-cHIS patients, cHIS patients were older, fewer female, more Blacks, higher prevalence of pre-existing comorbidities. Patients with cHIS had higher risk of developing cardiovascular disease (HR = 1.24 [1.04,1.47] p < 0.05), CKD (1.24 [1.01, 1.53] p < 0.05), and obesity (1.61 [1.31,1.98], p < 0.001) but not hypertension, diabetes, COPD, and asthma. Cumulative incidence analysis showed that patients ≥ 50 years old showed markedly higher new incidences of individual new disorders compared to patients < 50 years old. COVID-19 related hyperinflammatory syndrome confers a significantly higher risk for developing new common clinical disorders. Identifying risks for developing new clinical disorders in patients with COVID-19 related hyperinflammatory syndrome may encourage diligent follow-up of high-risk individuals.
Collapse
Affiliation(s)
- Kevin E Duong
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Justin Y Lu
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Stephen Wang
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Tim Q Duong
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
- Center for Health & Data Innovation, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
18
|
Oba S, Hosoya T, Kawata D, Komiya Y, Iwai H, Koike R, Miyamoto S, Kanno T, Ainai A, Suzuki T, Hasegawa H, Yasuda S. Iguratimod, a promising therapeutic agent for COVID-19 that attenuates excessive inflammation in mouse models. Eur J Pharmacol 2025; 996:177537. [PMID: 40147575 DOI: 10.1016/j.ejphar.2025.177537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
In severe COVID-19 patients, excessive inflammation can lead to multiorgan dysfunction. Current anti-inflammatory treatments like glucocorticoids partially improve the outcomes, while immune systems are compromised. We have identified that SARS-CoV-2-infected obese mice were a good model of the cytokine storm seen in COVID-19. Here, we revealed that iguratimod (IGU), an approved agent for rheumatoid arthritis, improved survival by attenuating inflammation with minimal immune suppression. In this study, C57BL/6 mice were fed a high-fat diet (HFD) or a normal-fat diet (NFD) for ten weeks before being infected with a mouse-adapted SARS-CoV-2. IGU significantly improved survival rates and reduced lung inflammation in HFD-fed mice, with minimal impact on interferon-induced genes and viral load. Meanwhile, dexamethasone (DEX) did not improve survival, while it suppressed various immune reactions with different mechanisms to IGU. Interestingly, IGU-treated mice had fewer SARS-CoV-2 positive cells in the lung, although viral replication was comparable to the control mice. Neither IGU nor DEX inhibited the SARS-CoV-2 infection in Vero-E6 cells, unlike the antiviral agent, remdesivir. Of note, IGU was effective prophylactically and therapeutically in HFD mice, and showed beneficial effects in NFD-fed mice with a lethal dose exposure of SARS-CoV-2. We demonstrated that IGU could be a promising treatment for severe COVID-19, especially in obese patients, by fine-tuning inflammation without compromising antiviral immunity. This study supports the possibility of drug repositioning for IGU COVID-19 beyond autoimmune diseases.
Collapse
Affiliation(s)
- Seiya Oba
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Tadashi Hosoya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan.
| | - Daisuke Kawata
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Yoji Komiya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Hideyuki Iwai
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Ryuji Koike
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Takayuki Kanno
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Hideki Hasegawa
- WHO Collaborating Centre for Reference and Research on Influenza, Tokyo, Japan; Research Center for Influenza and Respiratory Virus, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan.
| |
Collapse
|
19
|
Vaikath NN, Al-Nesf MA, Majbour N, Abdesselem HB, Gupta V, Bensmail I, Abdi IY, Elmagarmid KA, Shabani S, Sudhakaran IP, Ghanem SS, Al-Maadheed M, Mohamed-Ali V, Blackburn JM, Decock J, El-Agnaf OMA. In-house assays for detecting anti-SARS-CoV-2 antibodies in serum and urine: Correlation with COVID-19 severity from a cohort study in Qatar. J Infect Public Health 2025; 18:102744. [PMID: 40117875 DOI: 10.1016/j.jiph.2025.102744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Serological assays targeting antibodies against key viral proteins, including the Spike (S1), Receptor Binding Domain (RBD), and Nucleocapsid, play a critical role in understanding immunity and supporting diagnostic efforts during COVID-19 pandemic, and afterward. This study aimed to develop and validate in-house assays for detecting anti-SARS-CoV-2 antibodies in serum and urine. METHODS ELISA-based assay was developed to detect IgG and IgM antibodies against SARS-CoV-2. The assay was examined in serum and urine samples of two different cohort of patients affected by COVID-19 disease with different severity and compared to age and sex matched control group. Neutralizing antibody activity was evaluated using an RBD-ACE2 binding inhibition assay. Additionally, a Sengenics protein microarray platform was employed to assess epitope-specific antibody responses. RESULTS The in-house ELISA assay reliably detected antibodies in both 163 serum and 64 urine samples compared to 50 serum samples from healthy control, with strong correlations observed between antibody levels in the two biofluids. Neutralizing antibody levels correlated positively with disease severity, highlighting their clinical relevance. The performance of the in-house assays was comparable to commercial kits, and the Sengenics microarray provided detailed insights into antibody profiles, identifying dominant epitopes within the Nucleocapsid core domain and RBD. CONCLUSIONS The developed in-house assay demonstrated robust performance and versatility, offering a cost-effective and scalable alternative to commercial kits. Their ability to detect antibodies in both serum and urine highlighted their potential as non-invasive diagnostic tools. These findings contribute to advancing sero-diagnostic capabilities, improving understanding of immune responses to SARS-CoV-2, and supporting global efforts to monitor and manage COVID-19 effectively.
Collapse
Affiliation(s)
- Nishant N Vaikath
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Maryam Ali Al-Nesf
- Allergy and Immunology Division, Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar; Center of Metabolism and Inflammation, Division of Medicine, Royal Free Campus, University College London, Rowland Hill Road, London NW3 2PF, UK
| | - Nour Majbour
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar; Translational Medicine, Neuroscience, Pharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Houari B Abdesselem
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar; Proteomics Core Facility, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar; College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Vijay Gupta
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Ilham Bensmail
- Proteomics Core Facility, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Ilham Y Abdi
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Khalifa Ahmed Elmagarmid
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Shadah Shabani
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Indulekha P Sudhakaran
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Simona S Ghanem
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Mohammed Al-Maadheed
- Center of Metabolism and Inflammation, Division of Medicine, Royal Free Campus, University College London, Rowland Hill Road, London NW3 2PF, UK; Anti-Doping Laboratory Qatar, Doha, Qatar
| | - Vidya Mohamed-Ali
- Center of Metabolism and Inflammation, Division of Medicine, Royal Free Campus, University College London, Rowland Hill Road, London NW3 2PF, UK; Anti-Doping Laboratory Qatar, Doha, Qatar
| | - Jonathan M Blackburn
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, South Africa; Sengenics Corporation, Level M, Plaza Zurich, Damansara Heights, Kuala Lumpur 50490, Malaysia; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Julie Decock
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar; Cancer Research Center (CRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Omar M A El-Agnaf
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar; College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| |
Collapse
|
20
|
Wu X, Tang L, Huang W, Gao M, Xu C, Li P, Kong X. Membrane Protein of SARS-CoV-2 Promotes the Production of CXCL10 and Apoptosis of Myocardial Cells. Cardiovasc Toxicol 2025; 25:830-840. [PMID: 40293660 DOI: 10.1007/s12012-025-10001-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
SARS-CoV-2 infections directly or indirectly cause unconscionable vascular events, significantly increasing the morbidity and mortality of COVID-19. Biomarkers associated with cardiac injury often elevate in individuals with COVID-19. Cytokine storm is a mechanism underlying myocardial cell injury caused by SARS-CoV-2 infections. Cell apoptosis in AC16 cells overexpressing structural and helper proteins of SARS-CoV-2 was detected by Western blot, MTT and TUNEL assay. The M protein was determined to play the most pronounced role in inducing apoptosis. Transcriptome sequencing on AC16 cells overexpressing the M protein was performed to screen differentially expressed genes (DEGs), which were further subjected to the gene set enrichment analysis. The regulatory effect of CXCL10 on cell apoptosis of AC16 cells overexpressing M protein was finally explored. Overexpression of M protein significantly increased the Bax/Bcl-2 and cleaved caspase-3/caspase-3(CC3/C3) ratios and the percentage of TUNEL-positive cells in AC16 cells, while markedly reducing cell viability. CXCL10 was the most prominent DEG in AC16 cells overexpressing M protein. Knockdown of CXCL10 partially reversed the increases in the Bax/Bcl-2 and cleaved caspase-3/caspase-3 ratios, the percentage of TUNEL-positive cells, as well as the release of pro-inflammatory cytokines in AC16 cells overexpressing M protein. The M protein of SARS-CoV-2 triggers the production of CXCL10 and apoptosis of myocardial cells.
Collapse
Affiliation(s)
- Xiaoguang Wu
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Lu Tang
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Wen Huang
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Min Gao
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Changhao Xu
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China.
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
21
|
Monsalve DM, Acosta-Ampudia Y, Acosta NG, Celis-Andrade M, Şahin A, Yilmaz AM, Shoenfeld Y, Ramírez-Santana C. NETosis: A key player in autoimmunity, COVID-19, and long COVID. J Transl Autoimmun 2025; 10:100280. [PMID: 40071133 PMCID: PMC11894324 DOI: 10.1016/j.jtauto.2025.100280] [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: 02/04/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
NETosis, the process through which neutrophils release neutrophil extracellular traps (NETs), has emerged as a crucial mechanism in host defense and the pathogenesis of autoimmune responses. During the SARS-CoV-2 pandemic, this process received significant attention due to the central role of neutrophil recruitment and activation in infection control. However, elevated neutrophil levels and dysregulated NET formation have been linked to coagulopathy and endothelial damage, correlating with disease severity and poor prognosis in COVID-19. Moreover, it is known that SARS-CoV-2 can induce persistent low-grade systemic inflammation, known as long COVID, although the underlying causes remain unclear. It has been increasingly acknowledged that excessive NETosis and NET generation contribute to further pathophysiological abnormalities following SARS-CoV-2 infection. This review provides an updated overview of the role of NETosis in autoimmune diseases, but also the relationship between COVID-19 and long COVID with autoimmunity (e.g., latent and overt autoimmunity, molecular mimicry, epitope spreading) and NETosis (e.g., immune responses, NET markers). Finally, we discuss potential therapeutic strategies targeting dysregulated NETosis to mitigate the severe complications of COVID-19 and long COVID.
Collapse
Affiliation(s)
- Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Nicolás Guerrero Acosta
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mariana Celis-Andrade
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Ali Şahin
- Selcuk University, Faculty of Medicine, Konya, Turkiye
| | - Ahsen Morva Yilmaz
- TUBITAK Marmara Research Center (TUBITAK-MAM), Life Sciences, Medical Biotechnology Unit, Kocaeli, Turkiye
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Reichman University, Herzelia, Israel
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
22
|
Gheraibia S, Belattar N, Hassan ME, El-Nekeety AA, El-Sawy ER, Abdel-Wahhab MA. Molecular docking of polyphenol compounds and exploring the anticoagulant activity of Costus speciosus extracts in vitro and in vivo. Toxicol Rep 2025; 14:101961. [PMID: 40092046 PMCID: PMC11908605 DOI: 10.1016/j.toxrep.2025.101961] [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: 09/16/2024] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Anticoagulants have an important role in the prevention of cardiovascular disorders. Costus speciosus (Costaceae) is a medicinal herb used to treat COVID-19-induced thrombosis. The purpose of this study was to assess the anticoagulant activity of various C. speciosus aqueous (CSAE), ethanol (SCEE), and methanol (CSME) extracts in vivo and in vitro utilizing thrombin time (TT), activated partial thromboplastin time (aPTT), and prothrombin time (PT). Different concentrations of the three extracts were used to evaluate the anticoagulation effects in vitro. In the in vivo assay, male Sprague Dawley rats were used to test the CSME as in vivo anticoagulants. Three groups of rats included the control group and the groups that received CSME daily at a low (200 mg/kg) or high dose (400 mg/kg b.w) for 2 weeks. The molecular docking of the major bioactive constituents of the methanolic extract against the binding site of the thrombin inhibitor complex was evaluated. The HPLC detected 13, 10 and 11 polyphenols in the methanolic, ethanolic and aqueous extracts, respectively. The in vitro results showed that all the studied extracts had anticoagulant activity and increased aPTT, TT, and PT time. The in vivo experiment supported the in vitro results and demonstrated that CSME greatly prolonged the anticoagulant characteristics when compared to the negative control. Both findings suggested that these extracts have significant anticoagulant activity, with CSME being more effective and potentially useful in pharmaceutical applications as a natural anticoagulant medication.
Collapse
Affiliation(s)
- Sara Gheraibia
- Laboratory of Applied Biochemistry, Faculty of Sciences of Nature and Life, Ferhat Abbes University, Setif 1, Algeria
| | - Noureddine Belattar
- Laboratory of Applied Biochemistry, Faculty of Sciences of Nature and Life, Ferhat Abbes University, Setif 1, Algeria
| | - Marwa E. Hassan
- Toxicology Department, Research Institute of Medical Entomology, Giza, Egypt
| | - Aziza A. El-Nekeety
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt
| | - Eslam R. El-Sawy
- Chemistry of Natural Products Department, National Research Centre, Dokki, Cairo, Egypt
| | - Mosaad A. Abdel-Wahhab
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt
| |
Collapse
|
23
|
Anderson M, Lopez J, Wyr M, Ramirez PW. Defining diverse spike-receptor interactions involved in SARS-CoV-2 entry: Mechanisms and therapeutic opportunities. Virology 2025; 607:110507. [PMID: 40157321 DOI: 10.1016/j.virol.2025.110507] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is an enveloped RNA virus that caused the Coronavirus Disease 2019 (COVID-19) pandemic. The SARS-CoV-2 Spike glycoprotein binds to angiotensin converting enzyme 2 (ACE2) on host cells to facilitate viral entry. However, the presence of SARS-CoV-2 in nearly all human organs - including those with little or no ACE2 expression - suggests the involvement of alternative receptors. Recent studies have identified several cellular proteins and molecules that influence SARS-CoV-2 entry through ACE2-dependent, ACE2-independent, or inhibitory mechanisms. In this review, we explore how these alternative receptors were identified, their expression patterns and roles in viral entry, and their impact on SARS-CoV-2 infection. Additionally, we discuss therapeutic strategies aimed at disrupting these virus-receptor interactions to mitigate COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Michael Anderson
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Julian Lopez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Maya Wyr
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Peter W Ramirez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA.
| |
Collapse
|
24
|
Peng W, Wei X, Wu Y, Shi C, Liu X, Wu J, Yang H, Rong N, Zhao B, Zhang G, Zhang W, Liu J, Liu J, Yang J. Dynamic Molecular Changes in Brain, Lung, and Heart of Hamsters Infected With SARS-CoV-2: Insights From a Severe and Recovery Phase Model. J Med Virol 2025; 97:e70410. [PMID: 40432336 DOI: 10.1002/jmv.70410] [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: 01/07/2025] [Revised: 04/13/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025]
Abstract
The Global pandemic of coronavirus disease 2019 was initiated by the emergence of severe acute respiratory syndrome coronavirus 2. In addition to conventional pulmonary lesions, a range of neurological injury symptoms have been identified in clinical practice, but the aetiology of neurological disorders linked to SARS-CoV-2 infection remains poorly understood. Syrian hamsters, which are highly susceptible to SARS-CoV-2 infection, exhibit a disease phenotype similar to that observed in human COVID-19 patients. In this study, a hamster model of COVID-19 infection was used to analyze molecular changes in different tissues at various time points post infection with distinct strains using proteomic and phosphoproteomic approaches. Multi-omics analysis showed that SARS-COV-2 infection triggers sustained downregulation of the abundance and phosphorylation levels of neuronal and synapse-associated proteins in the brain, suggesting that neuronal damage persists even during the recovery period. Additionally, infections with SARS-CoV-2 may contribute to the onset of long-term symptoms of COVID-19 by impacting energy metabolism, neurotransmitter release, and synaptic transmission pathways. This study provides a comprehensive molecular profile of hamsters infected with different SARS-CoV-2 strains in different tissues, offering foundational insights into the pathogenic mechanisms of COVID-19.
Collapse
Grants
- This study was supported by the National Key R&D Program of China (2023YFC2507102), the CAMS Innovation Fund for Medical Sciences (CIFMS) grant (2022-I2M-1-020, 2022-12M-CoV19-002, 2022-I2M-2-001, 2022-I2M-1-011, 2021-I2M-1-057, 2021-I2M-1-049, 2021-I2M-1-044, 2021-I2M-1-016, 2021-I2M-1-001 and 2022-I2M-CoV19-003), the Haihe Laboratory of Cell Ecosystem Innovation Fund (22HHXBSS00008 and 22HHKYZX0034), State Key Laboratory Special Fund 2060204, and the National Natural Science Foundation of China (Grants 32070543 and 82341064).
Collapse
Affiliation(s)
- Wanjun Peng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Xiaohui Wei
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Yue Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
| | - Chunmei Shi
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
| | - Xiaoyan Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
| | - Jing Wu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Hekai Yang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Na Rong
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Binbin Zhao
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Gengxin Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Wei Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Jiangfeng Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Juntao Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Pathogen Biology, Beijing, China
| |
Collapse
|
25
|
Wu H, Liu Z, Li Y. Intestinal microbiota and respiratory system diseases: Relationships with three common respiratory virus infections. Microb Pathog 2025; 203:107500. [PMID: 40139334 DOI: 10.1016/j.micpath.2025.107500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
In recent years, the role of the intestinal microbiota in regulating host health and immune balance has attracted widespread attention. This study provides an in-depth analysis of the close relationship between the intestinal microbiota and respiratory system diseases, with a focus on three common respiratory virus infections, including respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and influenza virus. The research indicates that during RSV infection, there is a significant decrease in intestinal microbial diversity, suggesting the impact of the virus on the intestinal ecosystem. In SARS-CoV-2 infection, there are evident alterations in the intestinal microbiota, which are positively correlated with the severity of the disease. Similarly, influenza virus infection is associated with dysbiosis of the intestinal microbiota, and studies have shown that the application of specific probiotics exhibits beneficial effects against influenza virus infection. Further research indicates that the intestinal microbiota exerts a wide and profound impact on the occurrence and development of respiratory system diseases through various mechanisms, including modulation of the immune system and production of short-chain fatty acids (SCFAs). This article comprehensively analyzes these research advances, providing new perspectives and potential strategies for the prevention and treatment of future respiratory system diseases. This study not only deepens our understanding of the relationship between the intestinal microbiota and respiratory system diseases but also offers valuable insights for further exploring the role of host-microbiota interactions in the development of diseases.
Collapse
Affiliation(s)
- Haonan Wu
- Department of Respiratory, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China
| | - Ziyu Liu
- The First Hospital of Jilin University, Changchun, China.
| | - Yanan Li
- Department of Respiratory, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
26
|
Hatakeyama J, Nakamura K, Kanda N, Kawauchi A, Fujitani S, Oshima T, Kato H, Ota K, Kamijo H, Asahi T, Muto Y, Hori M, Iba A, Hosozawa M, Iso H. Long-term functional prognosis with tocilizumab in severe COVID-19 infection: A multicenter prospective observational study on mechanically ventilated ICU patients in the COVID-19 recovery study II. J Infect Chemother 2025; 31:102708. [PMID: 40250803 DOI: 10.1016/j.jiac.2025.102708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Tocilizumab, an IL-6 receptor antagonist, may prevent functional impairments in critically ill patients by attenuating the cytokine storm. This study investigated a potential effect of tocilizumab on preventing functional impairments in patients with severe coronavirus infection 2019 (COVID-19). METHODS In a multicenter prospective observational study, patients with COVID-19 ≥ 20 years requiring mechanical ventilation admitted to the intensive care unit between April 2021 and September 2021 and discharged alive were followed for one year. A self-administered questionnaire on sequelae and functional impairments was mailed in August 2022, and data were collected. A multivariate logistic regression was used to assess the impact of tocilizumab on physical function, mental health, and Long COVID. RESULTS Of 157 analyzed patients, 41 received tocilizumab. The tocilizumab group had more severe illness, but a lower prevalence of physical impairment (17.1 % vs. 23.3 %, p = 0.41) and mental disorders (19.5 % vs. 39.7 %, p = 0.009) than the non-tocilizumab group. The prevalence of Long COVID was higher in the tocilizumab group (92.7 % vs. 80.2 %, p = 0.06), whereas fatigue/malaise was significantly lower (19.5 % vs. 37.1 %, p = 0.039). Adjusted odds ratios (95 % confidence interval) for physical impairment, mental disorders, and Long COVID with tocilizumab were 0.70 (0.2-2.1), 0.40 (0.16-1.01), and 2.94 (0.7-12.3), respectively, with no significant difference. CONCLUSIONS Tocilizumab was associated with a lower prevalence of physical impairment and mental disorders at 1 year in patients with severe COVID-19. Furthermore, Long COVID had a weaker impact on physical and cognitive functions.
Collapse
Affiliation(s)
- Junji Hatakeyama
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kensuke Nakamura
- Department of Critical Care Medicine, Yokohama City University Hospital, Kanagawa, Japan; Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Ibaraki, Japan.
| | - Naoki Kanda
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Ibaraki, Japan; Division of General Internal Medicine, Jichi Medical University Hospital, Tochigi, Japan
| | - Akira Kawauchi
- Department of Critical Care and Emergency Medicine, Japanese Red Cross Maebashi Hospital, Gunma, Japan
| | - Shigeki Fujitani
- Department of Emergency Medicine and Critical Care Medicine, St. Marianna University, Kanagawa, Japan
| | - Taku Oshima
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, Kanagawa, Japan
| | - Kohei Ota
- Department of Emergency and Critical Care Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Hiroshi Kamijo
- Department of Emergency and Critical Care Medicine, Shinshu University School of Medicine, Nagano, Japan
| | - Tomohiro Asahi
- Department of Cardiology, Naha City Hospital, Okinawa, Japan
| | - Yoko Muto
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| | - Miyuki Hori
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| | - Arisa Iba
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| | - Mariko Hosozawa
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| | - Hiroyasu Iso
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| |
Collapse
|
27
|
Chen GS, Lee T, Tsang JL, Binnie A, McCarthy A, Cowan J, Archambault P, Lellouche F, Turgeon AF, Yoon J, Lamontagne F, McGeer A, Douglas J, Daley P, Fowler R, Maslove DM, Winston BW, Lee TC, Tran KC, Cheng MP, Vinh DC, Boyd JH, Walley KR, Singer J, Marshall JC, Russell JA. Machine Learning Accurately Predicts Need for Critical Care Support in Patients Admitted to Hospital for Community-Acquired Pneumonia. Crit Care Explor 2025; 7:e1262. [PMID: 40443788 PMCID: PMC12119046 DOI: 10.1097/cce.0000000000001262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025] Open
Abstract
OBJECTIVES Hospitalized community-acquired pneumonia (CAP) patients are admitted for ventilation, vasopressors, and renal replacement therapy (RRT). This study aimed to develop a machine learning (ML) model that predicts the need for such interventions and compare its accuracy to that of logistic regression (LR). DESIGN This retrospective observational study trained separate models using random-forest classifier (RFC), support vector machines (SVMs), Extreme Gradient Boosting (XGBoost), and multilayer perceptron (MLP) to predict three endpoints: eventual use of invasive ventilation, vasopressors, and RRT during hospitalization. RFC-based models were overall most accurate in a derivation COVID-19 CAP cohort and were validated in one COVID-19 CAP and two non-COVID-19 CAP cohorts. SETTING This study is part of the Community-Acquired Pneumonia: Toward InnoVAtive Treatment (CAPTIVATE) Research program. PATIENTS Two thousand four hundred twenty COVID-19 and 1909 non-COVID-19 CAP patients over 18 years old hospitalized and not needing invasive ventilation, vasopressors, and RRT on the day of admission were included. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Performance was evaluated with area under the receiver operating characteristic curve (AUROC) and accuracy. RFCs performed better than XGBoost, SVM, and MLP models. For comparison, we evaluated LR models in the same cohorts. AUROC was very high ranging from 0.74 to 0.95 in predicting ventilation, vasopressors, and RRT use in our derivation and validation cohorts. ML used and variables such as Fio2, Glasgow Coma Scale, and mean arterial pressure to predict ventilator, vasopressor use, creatinine, and potassium to predict RRT use. LR was less accurate than ML, with AUROC ranging 0.66 to 0.8. CONCLUSIONS A ML algorithm more accurately predicts need of invasive ventilation, vasopressors, or RRT in hospitalized non-COVID-19 CAP and COVID-19 patients than regression models and could augment clinician judgment for triage and care of hospitalized CAP patients.
Collapse
Affiliation(s)
| | - Terry Lee
- Centre for Advancing Health Outcomes, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer L.Y. Tsang
- Critical Care Medicine, Niagara Health Knowledge Institute, St Catharines, ON, Canada
- Critical Care Medicine, McMaster University, Hamilton, ON, Canada
| | - Alexandra Binnie
- Critical Care Department, William Osler Health System, Brampton, ON, Canada
- Critical Care Medicine, Algarve Biomedical Centre, Faro, Portugal
- Critical Care Medicine, Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | - Anne McCarthy
- Infectious Disease, Ottawa Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Juthaporn Cowan
- Infectious Disease, Ottawa Research Institute, University of Ottawa, Ottawa, ON, Canada
| | | | - Francois Lellouche
- CHU de Québec-Université Laval Research Center, Population Health and Optimal Health Practices Unit, Trauma- Emergency- Critical Care Medicine, Québec City, QC, Canada
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Alexis F. Turgeon
- CHU de Québec-Université Laval Research Center, Population Health and Optimal Health Practices Unit, Trauma- Emergency- Critical Care Medicine, Québec City, QC, Canada
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Jennifer Yoon
- Critical Care Medicine, Humber River Hospital, Toronto, ON, Canada
| | | | - Allison McGeer
- Mt. Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Josh Douglas
- Critical Care Medicine, Lion’s Gate Hospital, North Vancouver, BC, Canada
| | - Peter Daley
- Infectious Disease, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Robert Fowler
- Critical Care Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - David M. Maslove
- Department of Critical Care, Kingston General Hospital and Queen’s University, Kingston, ON, Canada
| | - Brent W. Winston
- Departments of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Foothills Medical Centre, University of Calgary, Calgary, AB, Canada
| | - Todd C. Lee
- Division of Infectious Disease, McGill University, Montreal, QC, Canada
| | - Karen C. Tran
- Division of General Internal Medicine, Vancouver General Hospital, Vancouver, BC, Canada
| | - Matthew P. Cheng
- Division of Infectious Disease, McGill University, Montreal, QC, Canada
| | - Donald C. Vinh
- Division of Infectious Disease, McGill University, Montreal, QC, Canada
| | - John H. Boyd
- Centre for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Critical Care Medicine, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Keith R. Walley
- Centre for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Critical Care Medicine, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Joel Singer
- Centre for Advancing Health Outcomes, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - John C. Marshall
- Department of Surgery, St. Michael’s Hospital, Toronto, ON, Canada
| | - James A. Russell
- Centre for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Critical Care Medicine, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
28
|
Liao Y, Zeng T, Guo X, Li X. Ferritin's role in infectious diseases: Exploring pathogenic mechanisms and clinical implications. New Microbes New Infect 2025; 65:101582. [PMID: 40230813 PMCID: PMC11995792 DOI: 10.1016/j.nmni.2025.101582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Ferritin, an iron storage protein, is crucial for maintaining iron metabolism balance throughout the body and serves as a key biomarker for evaluating the body's iron reserves. Reduced ferritin levels typically indicate iron deficiency, whereas elevated ferritin levels indicate an acute inflammatory response in infectious diseases. Recent research has established a significant link between elevated ferritin levels and disease severity and prognosis. The concept of hyperferritinemic syndrome has underscored ferritin's role as a pathogenic mediator. During infections, ferritin not only serves as a biomarker of inflammation but also exerts pro-inflammatory functions, which is a key factor in perpetuating the vicious pathogenic cycle. This review offers a comprehensive exploration of ferritin, covering its structural characteristics, regulatory mechanisms, and how diverse pathogens modulate ferritin. Understanding its pivotal role in infectious diseases is essential for identifying novel therapeutic prospects and enhancing disease management and prevention.
Collapse
Affiliation(s)
| | | | - Xiaoyan Guo
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xinhua Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| |
Collapse
|
29
|
Xu Z, Li EH, Liu J, Zhang YJ, Xiao R, Chen XZ, Zhong ZH, Tang XJ, Fu LJ, Zhang H, Bao MH, Qi HB, Chen GL, Ding YB. Postpartum hemorrhage emerges as a key outcome of maternal SARS-CoV-2 omicron variant infection surge across pregnancy trimesters. J Infect Public Health 2025; 18:102733. [PMID: 40073664 DOI: 10.1016/j.jiph.2025.102733] [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: 07/25/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Despite evidence showing changes in disease severity with the virus's evolution and vaccination efforts, the link between maternal, perinatal, and neonatal outcomes and SARS-CoV-2 infections during different pregnancy trimesters remains unclear, especially with the BA.5 and BF.7 Omicron subvariant surge in China in December 2022. This study investigates the correlation between maternal, perinatal, and neonatal outcomes and SARS-CoV-2 infection across various pregnancy trimesters. METHODS This prospective cohort study was conducted at two hospitals in southwest China, examining the clinical records and infection status of 2158 pregnant women registered between January 1, 2022, and September 30, 2023. Initially shielded from COVID-19, the population later experienced a significant infection surge. A comparative analysis evaluated maternal, perinatal, and neonatal outcomes between infected and uninfected subjects. Primary outcomes included pregnancy complications and premature births, while secondary outcomes encompassed cesarean sections, delivery complications, and neonatal outcomes. RESULTS Pregnant women infected with SARS-CoV-2 had higher incidence of placenta increta/percreta and postpartum hemorrhage compared to uninfected women. First trimester infections were associated with a lower incidence of intrahepatic cholestasis of pregnancy [aOR = 0.29, 95 % CI 0.13-0.63] but a higher incidence of preterm birth [aOR = 2.16, 95 % CI 1.25-3.71]. Third trimester infections increased the risk of postpartum hemorrhage [aOR = 2.74, 95 % CI 1.21-6.18]. CONCLUSION SARS-CoV-2 infection during pregnancy is linked to increased incidence of placenta increta/percreta and postpartum hemorrhage. First trimester infections are associated with higher incidence of premature birth and lower incidence of intrahepatic cholestasis of pregnancy, while third trimester infections are linked to higher incidence of postpartum hemorrhage.
Collapse
Affiliation(s)
- Zhou Xu
- Department of Obstetrics and Gynecology, Sichuan Jinxin Xinan Women and Children's Hospital, Chengdu, Sichuan 610011, China
| | - Er-Han Li
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Jia Liu
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Yong-Jia Zhang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Rui Xiao
- Department of Obstetrics and Gynecology, Sichuan Jinxin Xinan Women and Children's Hospital, Chengdu, Sichuan 610011, China
| | - Xin-Zhen Chen
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Zhao-Hui Zhong
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Xiao-Jun Tang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Li-Juan Fu
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China; Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Hua Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016 China
| | - Mei-Hua Bao
- Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Hong-Bo Qi
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Gong-Li Chen
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Yu-Bin Ding
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha 410219, China.
| |
Collapse
|
30
|
Awad NK. Organs on chips: fundamentals, bioengineering and applications. J Artif Organs 2025; 28:110-130. [PMID: 39134691 DOI: 10.1007/s10047-024-01460-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/12/2024] [Indexed: 05/16/2025]
Abstract
Human body constitutes unique biological system containing specific fluid mechanics and biomechanics. Traditional cell culture techniques of 2D and 3D do not recapitulate these specific natures of the human system. In addition, they lack the spatiotemporal conditions of representing the cells. Moreover, they do not enable the study of cell-cell interactions in multiple cell culture platforms. Therefore, establishing biological system of dynamic cell culture was of great interest. Organs on chips systems were fabricated proving their concept to mimic specific organs functions. Therefore, it paves the way for validating new drugs and establishes mechanisms of emerging diseases. It has played a key role in validating suitable vaccines for Coronavirus disease (COVID-19). Herein, the concept of organs on chips, fabrication methodology and their applications are discussed.
Collapse
Affiliation(s)
- Nasser K Awad
- Physical Chemistry Department, Advanced Materials Technology and Mineral Resources Research Institute, National Research Centre, Dokki, 12422, Cairo, Egypt.
| |
Collapse
|
31
|
Zhang M, Zhao L, Zeng P, Mu X, Zhao J. Inflammatory and pulmonary function characteristics of bronchial asthma induced by COVID‑19 infection. Biomed Rep 2025; 22:101. [PMID: 40322552 PMCID: PMC12046281 DOI: 10.3892/br.2025.1979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/07/2025] [Indexed: 05/08/2025] Open
Abstract
Bronchial asthma, a widely prevalent respiratory disease influencing individuals of all age groups worldwide, has been increasingly recognized as a global concern. While there exists a potentially heightened risk of severe coronavirus disease 2019 (COVID-19) in asthmatic patients, particularly those with non-allergic asthma, it is uncertain whether COVID-19 infection-induced bronchial asthma has its own unique clinical characteristics. The present study aimed to compare and analyze the pulmonary function and eosinophilic inflammation indices of patients with COVID-19 infection-induced bronchial asthma and those with typical bronchial asthma, and further deepen the understanding of COVID-19 infection-induced bronchial asthma. A retrospective analysis was conducted on the pulmonary function and inflammatory characteristics of 116 patients diagnosed with COVID-19 infection-induced bronchial asthma and treated in outpatient clinics after March 2023, as well as 113 patients with typical bronchial asthma diagnosed and treated from January 2022 to November 2022. The main clinical characteristics were cough, sputum, chest tightness, dyspnea and wheezing. There was no significant difference in clinical characteristics between the two groups. The results indicated that there was no significant difference in the total IgE, the absolute value and percentage of eosinophil, transoral FeNO, and trans-nasal FeNO in the peripheral blood samples of patients in the COVID-19 infection-induced bronchial asthma group compared with the typical bronchial asthma group. Although there was no significant difference between the two groups in the rates of impairment in ventilation function, reserve function, and small airway function, significant differences were identified in various indicators, including forced expiratory volume in 1 sec as a percentage of the predicted value (FEV1%), residual volume/total lung capacity (RV/TLC), peak expiratory flow (PEF), maximal expiratory flow rate at 75% (MEF75), maximal voluntary ventilation (MVV), FEV * 30, and residual volume (RV) between the two groups. The findings indicated that patients with COVID-19 infection-induced bronchial asthma exhibited a comparatively inferior pulmonary function versus those with typical bronchial asthma. However, it is important to note that the clinical impact of this disparity was not statistically significant.
Collapse
Affiliation(s)
- Mingqiang Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P.R. China
| | - Lina Zhao
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P.R. China
| | - Pu Zeng
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P.R. China
| | - Xiangdong Mu
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P.R. China
| | - Jingquan Zhao
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P.R. China
| |
Collapse
|
32
|
Iwasaki T, Arimura H, Inui S, Kodama T, Cui YH, Ninomiya K, Iwanaga H, Hayashi T, Abe O. Predictive models of severe disease in patients with COVID-19 pneumonia at an early stage on CT images using topological properties. Radiol Phys Technol 2025; 18:534-546. [PMID: 40293683 PMCID: PMC12103364 DOI: 10.1007/s12194-025-00906-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025]
Abstract
Prediction of severe disease (SVD) in patients with coronavirus disease (COVID-19) pneumonia at an early stage could allow for more appropriate triage and improve patient prognosis. Moreover, the visualization of the topological properties of COVID-19 pneumonia could help clinical physicians describe the reasons for their decisions. We aimed to construct predictive models of SVD in patients with COVID-19 pneumonia at an early stage on computed tomography (CT) images using SVD-specific features that can be visualized on accumulated Betti number (BN) maps. BN maps (b0 and b1 maps) were generated by calculating the BNs within a shifting kernel in a manner similar to a convolution. Accumulated BN maps were constructed by summing BN maps (b0 and b1 maps) derived from a range of multiple-threshold values. Topological features were computed as intrinsic topological properties of COVID-19 pneumonia from the accumulated BN maps. Predictive models of SVD were constructed with two feature selection methods and three machine learning models using nested fivefold cross-validation. The proposed model achieved an area under the receiver-operating characteristic curve of 0.854 and a sensitivity of 0.908 in a test fold. These results suggested that topological image features could characterize COVID-19 pneumonia at an early stage as SVD.
Collapse
Affiliation(s)
- Takahiro Iwasaki
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
- Department of Radiology, The University of Tokyo Hospital, Tokyo, Japan.
| | - Hidetaka Arimura
- Division of Medical Quantum Science, Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shohei Inui
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takumi Kodama
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yun Hao Cui
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenta Ninomiya
- Harry Perkins Institute of Medical Research, The University of Western Australia, Western Australia, Australia
| | - Hideyuki Iwanaga
- Division of Financial Strategy Management, The University of Tokyo Hospital, Tokyo, Japan
| | - Toshihiro Hayashi
- Department of Radiology, The University of Tokyo Hospital, Tokyo, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
33
|
Hieu NM, Takefuji Y. Comment on "Reassessing predictive modeling for emergency department return in COVID-19 patients". Am J Emerg Med 2025; 92:211-212. [PMID: 39824675 DOI: 10.1016/j.ajem.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/20/2025] Open
Affiliation(s)
- Nguyen Minh Hieu
- Faculty of Data Science, Musashino University, 3-3-3 Ariake Koto-ku, Tokyo 135-8181, Japan.
| | - Yoshiyasu Takefuji
- Faculty of Data Science, Musashino University, 3-3-3 Ariake Koto-ku, Tokyo 135-8181, Japan.
| |
Collapse
|
34
|
Sang JC, Musyoki SK, Injera WE, Karani LW, Maiyoh GK. Cytokine immune profiles among COVID 19 patients with different disease severities seeking treatment at Moi teaching and referral hospital, Kenya. Cytokine 2025; 190:156917. [PMID: 40117838 DOI: 10.1016/j.cyto.2025.156917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND COVID-19 manifests with a wide range of severities, from asymptomatic to critical conditions. Immunological profiles in patients positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may serve as early indicators of disease severity, aiding in prioritizing patient care. METHODOLOGY Archived patient plasma samples were retrieved from the Molecular Lab Bio-repository, ensuring equal representation of males, females, and various disease severities. Socio-demographic and disease severity data were obtained from patient health records. Levels of pro-inflammatory cytokines (interferon-gamma [IFN-γ], tumor necrosis factor-alpha [TNF-α], interleukin-2 [IL-2], and interleukin-17 [IL-17]) and anti-inflammatory cytokines (interleukin-4 [IL-4], interleukin-6 [IL-6], and interleukin-10 [IL-10]) were measured using the BD FACSCalibur flow cytometer. Data analysis involved comparing cytokine levels across different disease severities, with demographic data expressed as means ± standard deviation (SD). Statistical significance was set at P ≤ 0.05. FINDINGS The mean ages for males and females were 49.6 ± 22.7 and 48.4 ± 23.7, respectively. Mean ages for disease severity categories were 33 ± 19 (asymptomatic), 45.2 ± 21.5 (moderate), 56.8 ± 18.7 (severe), and 61.95 ± 22 (critical). Comorbidities were present in 25 % of patients, with cardiovascular disease (41 %) and pulmonary disease (31 %) being the most common. Predominant symptoms in critical patients included dyspnea (63 %) and myalgia (60 %), while rhinorrhea (46.2 %) and chest pain (45.7 %) were common in severe cases. Gastrointestinal symptoms were observed only in severe and critical groups. Levels of the pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-17) increased linearly with disease severity. Among anti-inflammatory cytokines, IL-6 and IL-10 levels also rose significantly with increasing severity. CONCLUSION Levels of TNF-α, IL-17, and IL-6 correlated with disease severity and may serve as prognostic biomarkers. Advanced age and underlying comorbidities were independently associated with higher disease severity.
Collapse
Affiliation(s)
- Jenniffer C Sang
- Department of Medical Laboratory Sciences, School of Health Sciences, Kisii University, Kisii, Kenya; Department of Laboratory and pathology Services, Moi Teaching and Referral Hospital, P.O. Box 3 - 30100, Eldoret, Kenya.
| | - Stanslaus K Musyoki
- Department of Medical Laboratory Sciences, School of Health Sciences, South Eastern Kenya University, P.O. Box 170-90200, Kitui, Kenya
| | - Wilfred E Injera
- Department of Medical Laboratory Sciences, School of Health Sciences, Alupe University, Busia, Kenya
| | - Lucy W Karani
- Department of Medical Laboratory Sciences, School of Health Sciences, Kisii University, Kisii, Kenya
| | - Geoffrey K Maiyoh
- Department of Biochemistry and Clinical Chemistry, School of Medicine, Moi University, P.O Box 4606-30100, Eldoret, Kenya.
| |
Collapse
|
35
|
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
|
36
|
Xu WT, An XB, Chen MJ, Ma J, Wang XQ, Yang JN, Wang Q, Wang DY, Wu Y, Zeng L, Qu Y, Zhao B, Ai J. A Gene Cluster of Mitochondrial Complexes Contributes to the Cognitive Decline of COVID-19 Infection. Mol Neurobiol 2025; 62:6869-6883. [PMID: 39271627 DOI: 10.1007/s12035-024-04471-3] [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: 04/14/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
"Brain fog," a persistent cognitive impairment syndrome, stands out as a significant neurological aftermath of coronavirus disease 2019 (COVID-19). Yet, the underlying mechanisms by which COVID-19 induces cognitive deficits remain elusive. In our study, we observed an upregulation in the expression of genes linked to the inflammatory response and oxidative stress, whereas genes associated with cognitive function were downregulated in the brains of patients infected with COVID-19. Through single-nucleus RNA sequencing (snRNA-seq) analysis, we found that COVID-19 infection triggers the immune responses in microglia and astrocytes and exacerbates oxidative stress in oligodendrocytes, oligodendrocyte progenitors (OPCs), and neurons. Further investigations revealed that COVID-19 infection elevates LUC7L2 expression, which inhibits mitochondrial oxidative phosphorylation (OXPHOS) and suppresses the expression of mitochondrial complex genes such as MT-ND1, MT-ND2, MT-ND3, MT-ND4L, MT-CYB, MT-CO3, and MT-ATP6. A holistic approach to protect mitochondrial complex function, rather than targeting a single molecular, should be an effective therapeutic strategy to prevent and treat the long-term consequences of "long COVID."
Collapse
Affiliation(s)
- Wen-Tao Xu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Xiao-Bin An
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Mei-Jie Chen
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Jing Ma
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Xu-Qiao Wang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Ji-Nan Yang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Qin Wang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Dong-Yang Wang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Yan Wu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Lu Zeng
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Yang Qu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China
| | - Bowen Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Jing Ai
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, Heilongjiang Province, Harbin, 150086, China.
| |
Collapse
|
37
|
Chen J, Zhao S, Yan H, Huang Y, Wei C, Liu J, Sun J. Plasma SARS-CoV-2 nucleocapsid antigen levels are associated with lung infection and tissue-damage biomarkers. Virus Res 2025; 356:199580. [PMID: 40339608 DOI: 10.1016/j.virusres.2025.199580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/17/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND SARS-CoV-2 nucleocapsid (N) antigen has been confirmed in the peripheral blood of patients with new coronavirus infection,yet its diagnostic and prognostic significance remains unclear. This study aimed to characterize the dynamics of SARS-CoV-2 N antigenemia in patients with novel coronavirus positivity, and to assess its potential association with clinical severity and plasma biomarker levels. METHODS We analyzed the level of SARS-CoV-2 N antigen, spike receptor-binding domain (S-RBD) IgG, neutralizing antibodies (NAb) and tissue-damage biomarkers was assessed in 180 plasma samples from 51 SARS-CoV-2-positive individuals. Plasma antigen levels were compared with concurrent respiratory nucleic acid amplification test results. RESULTS Patients with Ct values below 30 showed significantly different serum antigen levels compared to those with Ct values above 30 (p < 0.01). However, no significant positive correlation was found between respiratory viral load and serum antigen levels. Further analysis revealed that patients with pneumonia had markedly higher serum antigen levels than those without (p < 0.0001). Additionally, serum amyloid A (SAA) and ferritin (Fe) levels were significantly elevated in the antigenemia-positive group compared to the negative group, while procalcitonin (PCT) and interleukin-6 (IL-6) levels showed no significant differences. Notably, the positivity rate of N antigen in peripheral blood peaked at 47.1% (95% CI: 37.8%-56.7%) during the first week of infection and then gradually decreased over time. Moreover, patients with severe COVID-19 exhibited significantly higher serum antigen levels than those with mild or moderate disease (p < 0.0001). Serum levels of SARS-CoV-2 S-RBD IgG and neutralizing antibodies (NAb) were also significantly higher in antigenemia-negative patients than in antigenemia-positive patients (p < 0.0001). CONCLUSIONS Our findings highlight the multifaceted role of antigenemia in SARS-CoV-2 and suggest its potential as a biomarker for disease monitoring and risk stratification.
Collapse
Affiliation(s)
- Jing Chen
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Shuai Zhao
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Haiyang Yan
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Yaomeng Huang
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Congzhen Wei
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China
| | - Jiajia Liu
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China.
| | - Jingna Sun
- Department of Clinical Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Innovation Center of Clinical Medical Laboratory Technology, Shijiazhuang, Hebei, China.
| |
Collapse
|
38
|
Nègre P, Tayac D, Jamme T, Combis MS, Maupas-Schwalm F. Early suPAR levels as a predictor of COVID-19 severity: A new tool for efficient patient triage. Infect Dis Now 2025; 55:105058. [PMID: 40101896 DOI: 10.1016/j.idnow.2025.105058] [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: 11/07/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Following several waves of the COVID-19 pandemic, we are now facing a lower but persistent rate of SARS-CoV-2 infections, with seasonal resurgences often coinciding with other respiratory tract infections. OBJECTIVE We aimed to identify early clinico-biological variables predictive of an unfavorable outcome in patients with primary SARS-CoV-2 infection. We also evaluated the role of suPAR, an innovative biomarker, in predicting disease severity. METHODS We included 255 patients with PCR-confirmed primary SARS-CoV-2 infection and with a 30-day follow-up minimum. Blood samples were collected within the first 24 h of hospitalization to measure suPAR levels. Comprehensive data from medical records were analyzed to assess their predictive value in stratifying patients into seven severity groups, with groups 1 to 3 representing severe COVID-19 (death, intubation, ECMO, or non-invasive ventilation). RESULTS Early plasma suPAR levels were significantly associated with severe disease progression, as evidenced by ANOVA and logistic regression models, highlighting suPAR as a persistent predictive factor for unfavorable outcomes. CONCLUSION Our findings suggest that a single suPAR measurement, performed early after a positive PCR test for SARS-CoV-2, holds strong predictive value for patient outcomes. This biomarker, alongside pulse oximetry and CT scan results, could be instrumental in early patient triage during seasonal COVID-19 resurgences.
Collapse
Affiliation(s)
- Pauline Nègre
- Faculty of Pharmacy, Toulouse III University, France; Medical Biochemistry Laboratory, CHU Toulouse, France
| | - Didier Tayac
- Medical Biochemistry Laboratory, CHU Toulouse, France
| | - Thibaut Jamme
- Medical Biochemistry Laboratory, CHU Toulouse, France
| | | | - Françoise Maupas-Schwalm
- Medical Biochemistry Laboratory, CHU Toulouse, France; Faculty of Medicine, Toulouse III University, France.
| |
Collapse
|
39
|
Piulachs X, Langohr K, Besalú M, Pallarès N, Carratalà J, Tebé C, Melis GG. Semi-Markov Multistate Modeling Approaches for Multicohort Event History Data. Biom J 2025; 67:e70051. [PMID: 40342140 DOI: 10.1002/bimj.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 10/22/2024] [Accepted: 01/28/2025] [Indexed: 05/11/2025]
Abstract
Two Cox-based multistate modeling approaches are compared for modeling a complex multicohort event history process. The first approach incorporates cohort information as a fixed covariate, thereby providing a direct estimation of the cohort-specific effects. The second approach includes the cohort as a stratum variable, which offers an extra flexibility in estimating the transition probabilities. Additionally, both approaches may include possible interaction terms between the cohort and a given prognostic predictor. Furthermore, the Markov property conditional on observed prognostic covariates is assessed using a global score test. Whenever departures from the Markovian assumption are revealed for a given transition, the time of entry into the current state is incorporated as a fixed covariate, yielding a semi-Markov process. The two proposed methods are applied to a three-wave dataset of COVID-19-hospitalized adults in the southern Barcelona metropolitan area (Spain), and the corresponding performance is discussed. While both semi-Markovian approaches are shown to be useful, the preferred one will depend on the focus of the inference. To summarize, the cohort-covariate approach enables an insightful discussion on the behavior of the cohort effects, whereas the stratum-cohort approach provides flexibility to estimate transition-specific underlying risks according to the different cohorts.
Collapse
Affiliation(s)
- Xavier Piulachs
- Department of Statistics and Operations Research, Polytechnic University of Catalonia, Barcelona, Spain
| | - Klaus Langohr
- Department of Statistics and Operations Research, Polytechnic University of Catalonia, Barcelona, Spain
| | - Mireia Besalú
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Natàlia Pallarès
- Bellvitge Biomedical Research Institute, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Carratalà
- Bellvitge Biomedical Research Institute, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Infectious Diseases, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Center for Biomedical Research in Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Cristian Tebé
- Bellvitge Biomedical Research Institute, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Guadalupe Gómez Melis
- Department of Statistics and Operations Research, Polytechnic University of Catalonia, Barcelona, Spain
| |
Collapse
|
40
|
Scheinberg P, Khoshnevis MR, Robinson PA, Guerreros A, Sato VAH, Fonseca BAL, Prozesky HW, Romero JOC, Fogliatto L, Meisenberg BR, Park DJ, Gupta A, Patel P, Townsley DM, Zheng L, Munugalavadla V. Efficacy and safety of acalabrutinib with best supportive care versus best supportive care in patients with COVID-19 requiring hospitalization. Immunohorizons 2025; 9:vlaf023. [PMID: 40461100 PMCID: PMC12133263 DOI: 10.1093/immhor/vlaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 06/11/2025] Open
Abstract
The efficacy and safety of acalabrutinib, a Bruton tyrosine kinase (BTK) inhibitor, was evaluated in 2 phase 2 studies in hospitalized patients with coronavirus disease 2019 (COVID-19) who received acalabrutinib + best supportive care (BSC) versus BSC alone (Clinicaltrials.gov: NCT04380688 and NCT04346199). The primary endpoint was the percentage of patients alive and free of respiratory failure on day 14 (rest of the world [RoW] study) and day 28 (US study). In the RoW study, 177 patients were randomized (acalabrutinib + BSC: n = 89; BSC: n = 88); in the US study, 62 patients were randomized (acalabrutinib + BSC: n = 31; BSC: n = 31). The percentage of patients who met the primary endpoint was similar in both studies (RoW study: acalabrutinib + BSC: 83.1%, BSC: 90.9%; US study: acalabrutinib + BSC: 80.6%, BSC: 83.9%). No new safety concerns were reported. Overall, no significant clinical benefit of adding acalabrutinib to BSC in patients hospitalized with COVID-19 was observed.
Collapse
Affiliation(s)
- Phillip Scheinberg
- Division of Hematology, Hospital A Beneficência Portuguesa, São Paulo, Brazil
| | - Matt R Khoshnevis
- Center for Critical Care, Houston Methodist Hospital, Houston, TX, United States
| | - Philip A Robinson
- Infection Prevention and Hospital Epidemiology, Hoag Memorial Hospital Presbyterian, Newport Beach, CA, United States
| | | | - Victor A H Sato
- International Research Center, Hospital Alemao Oswaldo Cruz, São Paulo, Brazil
| | - Benedito A L Fonseca
- Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP), São Paulo, Brazil
| | - Hans W Prozesky
- Division of Infectious Diseases, Department of Medicine, Stellenbosch University and TREAD Research, Tygerberg Hospital, Cape Town, South Africa
| | - José Omar Chacón Romero
- Hospital Sub-direction, IMSS HGR No. 1 Dr Carlos Mac Gregor Sanchez Navarro, Mexico City, Mexico
| | - Laura Fogliatto
- Hematology and Hemotherapy Department, Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil
| | - Barry R Meisenberg
- Department of Medicine, Anne Arundel Medical Center, Annapolis, MD, United States
| | - David J Park
- Hematology and Oncology, Providence St Jude Medical Center/Providence Medical Foundation, Fullerton, CA, United States
| | - Ashok Gupta
- Oncology Research and Development, AstraZeneca, Gaithersburg, MD, United States
| | - Priti Patel
- Haematology Research and Development, AstraZeneca, South San Francisco, CA, United States
| | - Danielle M Townsley
- Oncology Research and Development, AstraZeneca, Gaithersburg, MD, United States
| | - Lianqing Zheng
- Oncology Biometrics, AstraZeneca, South San Francisco, CA, United States
| | | |
Collapse
|
41
|
Zabiegala A, Kim Y, Chang KO. Host susceptibilities and entry processes of SARS-CoV-2 Omicron variants using pseudotyped viruses carrying spike protein. BMC Vet Res 2025; 21:377. [PMID: 40426227 PMCID: PMC12108000 DOI: 10.1186/s12917-025-04822-9] [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: 02/17/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The zoonotic potential has been well studied for SARS-CoV-2 and its earlier variants, but the information for Omicron variants and SARS-CoV is lacking. In this study, we generated lentivirus-based pseudoviruses carrying spike protein (S) of SARS-CoV-2, parental and Omicron variants including BA.1.1, BA.4/5, XBB.1 and JN.1 to assess the entry into cells expressing human or animal ACE2 including dogs, cats and white-tailed deer. Using these pseudoviruses, along with pseudoviruses carrying S of MERS-CoV and SARS-CoV, we assessed the protease processing of these various S through western blotting, entry/inhibition assays, and fusion assays. The results showed that overall, pseudotyped viruses carrying each S of SARS-CoV-2 Omicron strains efficiently entered cells expressing human or animal ACE2 comparably (BA.1.1 and JN.1) or better (BA.4/5 and XBB.1) than those with parental strain. In addition, the entries of pseudotyped viruses carrying S of SARS-CoV were also efficient the cells expressing human or animal ACE2. The presence of TMPRSS2 significantly increased the entry of all tested pseudoviruses including those with S of MERS-CoV, SARS-CoV and SARS-CoV-2, with BA.1.1, JN1, and XBB.1 Omicron having the largest fold increase. When cathepsin inhibitors were examined to assess their inhibitory effects on entry of parental and Omicron variants, they were significantly less effective in the entry of Omicron variants compared to parent strain, suggesting Omicron strains do not depend on the endosomal route compared to parental strain.
Collapse
Affiliation(s)
- Alexandria Zabiegala
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS, 66506, USA
| | - Yunjeong Kim
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS, 66506, USA
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS, 66506, USA.
| |
Collapse
|
42
|
Dias JDS, Rebouças MAM, Carvalho LVDS, Silva TS, Santos JSD, Melendez AXTO, Brites C. Neurological manifestations of acute SARS-CoV-2 infection in a reference hospital in Bahia, Brazil. Braz J Infect Dis 2025; 29:104542. [PMID: 40424700 DOI: 10.1016/j.bjid.2025.104542] [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: 12/26/2024] [Revised: 03/10/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Neurologic manifestations of Coronavirus Disease-19 (COVID-19) have been associated with patients' disease severity and outcome. This study aimed to describe the frequency and characteristics of the neurological manifestations in a group of hospitalized individuals with COVID-19 and their associations with patient outcomes. METHODS Patients aged 18 years or older admitted to a local hospital between April and June 2020 with SARS-CoV-2 detected by RT-PCR were included in this retrospective observational study. The characteristics of participants were collected from electronic medical records using a structured questionnaire. A Poisson regression model was used to examine the influence of neurological manifestations on mortality. RESULTS A total of 305 participants with COVID-19 were included, with 57.7 % of them presenting neurological symptoms. There were 62 (20.3 %) individuals with acute encephalopathy, with a mean age of 65.5 ± 15.9 years. In this group, higher Prevalence Ratios (PR) of comorbidities (1.6) and severe disease (3.6) were present, predisposing factors for acute encephalopathy. They were also more likely to be admitted to the intensive care unit (3.1) and to die (2.4). The median Neutrophil-Lymphocyte Ratio (NLR) was 7 (Interquartile Range [IQR: 4‒12]). Fifty-two (17 %) participants presented chemosensory dysfunction, with a mean age 53.3 ± 14 years and a lower PR of comorbidity (0.8) than those without. The severe diseases' PR was slightly higher (1.1), but the PR of ICU admission (0.7), and deaths (0.4) was lower. The LNR was 3.8 (IQR: 2.2-7.8). Poisson regression analysis revealed that severe illness (PR = 3.13), cardiopathy (PR = 1.65), acute encephalopathy (PR = 1.49), diabetes (PR = 1.46), and neutrophil-lymphocyte ratio (PR = 1.04) were associated with death. Conversely, having chemosensory disorders (PR = 0.44) and a prolonged hospital stay (PR = 0.96) were associated with survival. CONCLUSION Patients with acute encephalopathy had more severe forms of COVID-19 and higher mortality. In contrast, chemosensory dysfunction was associated with milder disease manifestations and a better prognosis.
Collapse
Affiliation(s)
- Jesângeli de Sousa Dias
- Programa de Pós-Graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Instituto Couto Maia, Salvador, Bahia, Brazil.
| | | | | | | | | | | | - Carlos Brites
- Programa de Pós-Graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
43
|
Alkharaan H. Infectious and Immunological Links Between Periodontitis and COVID-19: A Review. Med Sci Monit 2025; 31:e948069. [PMID: 40418682 PMCID: PMC12124155 DOI: 10.12659/msm.948069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/01/2025] [Indexed: 05/28/2025] Open
Abstract
Emerging evidence suggests a potential association between periodontitis and adverse outcomes in COVID-19. Both conditions share risk factors and exhibit similar immune dysregulation, including elevated pro-inflammatory cytokines, altered myeloid compartments, and T-cell dysfunction. SARS-CoV-2 uses angiotensin-converting enzyme type 2 and transmembrane protease serine 2 membrane proteins, highly expressed in the oral cavity, for cellular entry. Periodontitis may exacerbate COVID-19 through mechanisms such as oral microbe aspiration, increased viral receptor expression, and systemic inflammation. The shared immunopathogenesis, characterized by cytokine storms and perturbed immune profiles, suggests periodontitis can predispose patients to more severe COVID-19 outcomes. This article aims to review the associations between periodontitis and the severity of COVID-19 and the possible immune mechanisms involved.
Collapse
|
44
|
Bucher AM, Frodl E, Ehrengut C, Müller L, Emrich T, Kloeckner R, Sieren M, Sähn MJ, Fink MA, Móré D, Melekh B, Gottschling S, Meinel F, Schön H, Kornemann N, Renz DM, Lubina N, Wollny C, May MS, Siegler L, Borggrefe J, Meyer HJ, Surov A. Prognostic value of CT-defined coronary sclerosis in COVID-19: results of a multicenter study based on the Weston score. ROFO-FORTSCHR RONTG 2025. [PMID: 40418966 DOI: 10.1055/a-2583-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Coronary calcification, as defined by computed tomography (CT), can be quantified with a score (CAC score). It is an established prognostic and predictive imaging marker of the cardiovascular risk profile. The prognostic relevance of the CAC score has been demonstrated for acute diseases, including the coronavirus disease 2019 (COVID-19) in preliminary studies. The aim of the present study was to prove the prognostic relevance of the CAC score in patients with coronavirus disease 2019.The present study used a nationwide radiological research platform to conduct a multicenter retrospective study. The study included a total of 541 patients, 176 of whom were female (32.5%). The mean age of the patients was 61.2 years ± 15.6 years. The coronavirus (SARS-COV-2) disease was confirmed in all patients by PCR testing. The CAC score was calculated using the Weston score, which is a semiquantitative method. The primary outcome of the study was 30-day mortality.The overall mortality rate within the 30-day period was 21.2%, with 115 patients dying. The mean Weston score was 3.0 ± 3.6. 128 patients (23.7%) exhibited no evidence of coronary calcifications, as indicated by a Weston Score of 0. In the univariable regression analysis, the presence of calcifications was found to be associated with mortality, with an odds ratio of 1.68 (95% confidence interval 1.08-2.59, p=0.01). However, this result did not remain statistically significant in the multivariable analysis (p=0.49). The Weston score was found to be associated with mortality in the univariable analysis, with an odds ratio (OR) of 1.10 (95% CI 1.04-1.14, p < 0.001), and in the multivariable analysis, with an OR of 1.06 (95% CI 1.005-1.138, p = 0.036).The imaging marker CAC score has been demonstrated to have a significant prognostic impact on 30-day mortality in patients diagnosed with coronavirus disease 2019 (COVID-19). It is incumbent upon the radiologist to acknowledge the sole presence of coronary calcifications as a relevant prognostic factor. The prognostic relevance of the calcifications was found to be greater in cases where more extensive calcification was present. · Coronary calcifications assessed by CT are prognostic markers for 30-day COVID-19 mortality.. · Calcification extent has greater prognostic value than the mere presence of calcifications.. · The study highlights the need to integrate coronary calcifications into clinical risk scores.. · Bucher AM, Frodl E, Ehrengut C et al. Prognostic value of CT-defined coronary sclerosis in COVID-19: results of a multicenter study based on the Weston score. Rofo 2025; DOI 10.1055/a-2583-0235.
Collapse
Affiliation(s)
- Andreas Michael Bucher
- University Hospital, Institute for Diagnostic and Interventional Radiology, Goethe University Frankfurt Faculty 16 Medicine, Frankfurt am Main, Germany
| | - Eric Frodl
- University Hospital, Institute for Diagnostic and Interventional Radiology, Goethe University Frankfurt Faculty 16 Medicine, Frankfurt am Main, Germany
| | | | - Lukas Müller
- Department for Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany
| | - Tilman Emrich
- Department for Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany
| | - Roman Kloeckner
- Department of Radiology, University Hospital Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Malte Sieren
- Department of Radiology, University Hospital Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Marwin-Jonathan Sähn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Matthias A Fink
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dorottya Móré
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Bohdan Melekh
- Department of Radiology and Nuclear Medicine, University Hospital of Magdeburg, Magdeburg, Germany
| | - Sebastian Gottschling
- Department of Radiology and Nuclear Medicine, University Hospital of Magdeburg, Magdeburg, Germany
| | - Felix Meinel
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Hospital of Rostock, Rostock, Germany
| | - Hanna Schön
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Hospital of Rostock, Rostock, Germany
| | - Norman Kornemann
- Department of Radiology, Hannover Medical School, Hanover, Germany
| | | | - Nora Lubina
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Claudia Wollny
- Clinic for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | | | - Lisa Siegler
- Department of Radiology, University Hospital of Erlangen, Erlangen, Germany
| | - Jan Borggrefe
- Institute of Radiology, Neuroradiology and Nuclear Medicine Minden, Ruhr-University-Bochum, Minden, Germany
| | - Hans-Jonas Meyer
- Department of Radiology, University Hospital Leipzig, Leipzig, Germany
| | - Alexey Surov
- Institute of Radiology, Neuroradiology and Nuclear Medicine Minden, Ruhr-University-Bochum, Minden, Germany
| |
Collapse
|
45
|
Mahesh T, Changela S, Duong KS, Henry S, Wang SH, Duong TQ. New-onset conjunctivitis 3.5 years post SARS-CoV-2 infection in an inner-city population in the Bronx. BMJ Open Ophthalmol 2025; 10:e001993. [PMID: 40404339 DOI: 10.1136/bmjophth-2024-001993] [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: 10/21/2024] [Accepted: 03/12/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND A few studies have reported conjunctivitis is a complication associated with acute COVID-19. It is unknown whether SARS-CoV-2 infection increases the risk of conjunctivitis post-COVID-19 long term. OBJECTIVES This study investigated the incidence of new-onset conjunctivitis 3.5 years post SARS-CoV-2 infection and compared it with patients without SARS-CoV-2 infection. METHODS This retrospective study consisted of 67 702 patients who tested positive for COVID-19 (defined by a positive PCR test), and 1 391 135 COVID-19-negative patients with no prior records of conjunctivitis in the Montefiore Health System from 11 March 2020 to 31 December 2022. The study included adult patients re-presenting to our centre with conjunctivitis. Outcome was new conjunctivitis between 14 days and 3.5 years post index date. Analysis was performed with unmatched and matched cohorts. Matching was done for age, sex, race and ethnicity. Cumulative incidence and hazard ratio (HR) with and without adjustment for competitive risks were analysed. RESULTS There were 1154 (2.27%) individuals with COVID-19 and contemporary 13 899 (1.57%) controls who developed new conjunctivitis. COVID-19-positive patients had a significantly higher risk of developing new incident conjunctivitis (unmatched cohort adjusted HR 1.11 (95% CI 1.04 to 1.17), matched cohort adjusted HR 1.10 (95% CI 1.02 to 1.16)) compared with COVID-19-negative patients. CONCLUSIONS COVID-19-positive patients had significantly higher risk of developing new conjunctivitis compared with contemporary COVID-19-negative controls. Identifying risk factors for developing new-onset conjunctivitis may draw clinical attention for careful follow-up in at-risk individuals for ocular infections.
Collapse
Affiliation(s)
- Tanya Mahesh
- Department of Radiology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sagar Changela
- Department of Radiology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Katie S Duong
- Department of Radiology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sonya Henry
- Department of Radiology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Stephen H Wang
- Department of Radiology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Tim Q Duong
- Department of Radiology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
46
|
Partouche N, Maumy M, Chamaraux-Tran TN, Bertrand F, Schneider F, Meyer N, Solis M, Fafi-Kremer S, Noll E, Pottecher J. Does the IL-6/KL-6 ratio distinguish different phenotypes in COVID-19 Acute Respiratory Distress Syndrome? An observational study stemmed from prospectively derived clinical, biological, and computed tomographic data. PLoS One 2025; 20:e0321533. [PMID: 40397955 DOI: 10.1371/journal.pone.0321533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 03/07/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND As new SARS-CoV-2 variants emerge and as treatment of COVID-19 ARDS remains exclusively supportive, there is an unmet need to better characterize its different phenotypes to tailor personalized treatments. Clinical, biological, spirometric and CT data hardly allow deciphering of Heavy (H), Intermediate (I) and Light (L) phenotypes of COVID-19 ARDS and the implementation of tailored specific strategies (prone positioning, PEEP settings, recruitment maneuvers). We hypothesized that the ratio of two pivotal COVID-19 biomarkers (interleukin 6 [IL-6] and Krebs von den Lungen 6 [KL-6], related to inflammation and pneumocyte repair, respectively) would provide a biologic insight into the disease timeline allowing 1) to differentiate H, I and L phenotypes, 2) to predict outcome and 3) to reflect some of CT findings. METHODS AND FINDINGS This was a retrospective analysis of prospectively acquired data (COVID HUS cohort). Inclusion concerned any patient with severe COVID-19 pneumonia admitted to two intensive care units between March 1st and May 1st, 2020, in a high-density cluster of the first epidemic wave (Strasbourg University Hospital, France). Demographic, clinical, biological (standard, IL-6 [new generation ELISA], KL-6 [CLEIA technique]), spirometric (driving pressure, respiratory system compliance) and CT data were collected longitudinally. CT analysis included semi-automatic and automatic lung measurements and allowed segmentation of lung volumes into 4 (poorly aerated, non-aerated, overinflated and normally aerated) and 3 (ground-glass, restricted normally aerated, and overinflated) zones, respectively. The primary outcome was to challenge the IL-6/KL-6 ratio capacity to decipher the three COVID-19 ARDS phenotypes (H, I and L) defined on clinical, spirometric and radiologic grounds. Secondary outcomes were the analysis of the prognostic value of the IL-6/KL-6 ratio and its correlates with CT-acquired data. Multivariate analysis was based on principal component analysis. One hundred and forty-eight ventilated COVID-19 ICU patients from the COVID HUS cohort were assessed for eligibility and 77 were included in the full analysis. Most were male, all were under invasive mechanical ventilation and vasopressor therapy and displayed high severity scores (SAPSII: 48 [42-56]; SOFA: 8 [7-10]). The L, I and H COVID ARDS phenotypes were identified in 11, 15 and 48 patients, respectively. In three patients, the phenotype could not be defined precisely. Thirty patients (39%) died in the ICU and the number of ventilator-free days was 2 [0-2] days. The IL-6/KL-6 ratio was not significantly different between the L, I and H phenotypes and evolved according to similar patterns over time. Surviving and deceased patients displayed an inverse kinetic of KL-6. IL-6 and the IL-6/KL-6 ratio were linearly associated with ground-glass volume on semi-automatic and automatic CT lung measurements. CONCLUSIONS In our population of severe ventilated COVID ARDS patients, the IL-6/KL-6 ratio was not clue to differentiate the H, I and L phenotypes and tailor a personalized ventilatory approach. There was an interesting correlation between IL-6/KL-6 ratio and ground-glass volume as determined by automated lung CT analysis. Such correlation deserves more in-depth pathophysiological study, at best gathered from a prospective cohort with a larger sample size and histological analysis. TRIAL REGISTRATION COVID HUS Trial registration number: NCT04405726.
Collapse
Affiliation(s)
- Nicolas Partouche
- Service d'Anesthésie-Réanimation & Médecine Péri-Opératoire, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, UR3072, FMTS, FHU Omicare, Faculté de Médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, Strasbourg, France
| | - Myriam Maumy
- LIST3N, University of Technology of Troyes, Troyes, France
| | - Thien-Nga Chamaraux-Tran
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 1 Rue Laurent Fries, Illkirch-Graffenstaden, France
| | | | - Francis Schneider
- Service de Médecine Intensive-Réanimation, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Nicolas Meyer
- Service de santé Publique, GMRC, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Morgane Solis
- Faculté de Médecine, Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France - INSERM, UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Samira Fafi-Kremer
- Faculté de Médecine, Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France - INSERM, UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Eric Noll
- Service d'Anesthésie-Réanimation & Médecine Péri-Opératoire, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, UR3072, FMTS, FHU Omicare, Faculté de Médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, Strasbourg, France
| | - Julien Pottecher
- Service d'Anesthésie-Réanimation & Médecine Péri-Opératoire, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, UR3072, FMTS, FHU Omicare, Faculté de Médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
47
|
Klimova EM, Lavinska OV, Drozdova LA. The role of DAMP cytotoxic fractions in the immune markers' disruption in patients with urgent surgical pathology and against the background of post-COVID-19 syndrome. Immunol Lett 2025:107033. [PMID: 40409597 DOI: 10.1016/j.imlet.2025.107033] [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: 12/20/2024] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND As a result of the SARS-CoV-2 pandemic, various population groups were formed that had acute and asymptomatic COVID-19. A survey in these groups revealed with equal frequency an asthenic symptom complex, the so-called post-COVID-19 syndrome (PCS). The frequency of urgent surgical pathology against the background of PCS and structural and functional disorders of various organs was increased. The aim - to study the dynamics of immunoresistance factors changes in patients with urgent surgical pathology that developed against the background of PCS and to identify pathogenic markers of the severe course and the risk of mortality. MATERIALS AND METHODS To examine patients with PCS and urgent cardiovascular (n = 103) and abdominal (n = 106) pathology we used the following methods: fluorescence microscopy, confocal microscopy, flow cytometry, spectrophotometry, ELISA. RESULTS We revealed a temporal dependence of immune dysfunction in patients with a comorbid course of urgent surgical pathology and PCS. The nature of the DAMP (damage-associated molecular patterns) cytotoxic fractions ratio was associated with certain changes in innate and adaptive immunity factors, severity of the condition and risk of mortality. At the first stage (2020-2021), patients with PCS has disorders of the humoral and cellular components of innate immunity against the background of an increase in the oligopeptide and peptide DAMP fractions. At the second stage (2022-2024) of PCS development, changes in innate as well as adaptive immunity were observed against the background of an increase in the cytotoxic oligonucleotide DAMP fraction (mortality was 17.3%). CONCLUSIONS The identified markers of impaired immunoresistance in cardiovascular and abdominal urgent pathology can be used to select targeted therapy tactics.
Collapse
Affiliation(s)
- Elena M Klimova
- Department of Molecular Biology and Biotechnology, V.N. Karazin Kharkiv National University, 4 Pl. Svobody, 61022 Kharkiv, Ukraine; Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine.
| | - Olena V Lavinska
- Department of Molecular Biology and Biotechnology, V.N. Karazin Kharkiv National University, 4 Pl. Svobody, 61022 Kharkiv, Ukraine; Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine
| | - Larisa A Drozdova
- Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine
| |
Collapse
|
48
|
Tizazu AM, Gize A, Ali S. Age influences blood cell-based immune deregulation antibody response and unfavorable clinical outcomes in COVID-19 patients. Sci Rep 2025; 15:17431. [PMID: 40394243 PMCID: PMC12092784 DOI: 10.1038/s41598-025-95722-3] [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: 09/30/2024] [Accepted: 03/24/2025] [Indexed: 05/22/2025] Open
Abstract
COVID-19 is caused by SARS-CoV-2 and has a diverse spectrum of clinical presentations ranging from asymptomatic cases to severe and critical cases that result in the death of the patient. Alongside the viral factors host factors like Age, deregulation of the immune response and presence of comorbidity determine the patient's outcome. Here we sought to assess the impact of age on natural antibody response, CBC-based inflammatory markers, and outcome of COVID-19 patients. We divided the participants into three groups, young (≤ 35 years), middle-aged (40-60 years), and old (≥ 65 years) patients and collected and analyzed sociodemographic, clinical, and laboratory parameters. We found that elderly patients showed higher (P < 0.05) levels of inflammation like increased neutrophil percentages, NLR, lymphopenia, and low Hgb levels, compared to middle-aged and young patients. Interestingly these markers were also associated with mortality of COVID-19 patients. On the other hand, no significant difference was observed in ion concentration, lipid profile, and coagulation test between the three age groups. We also found that elderly patients showed significantly (P < 0.05) decreased levels of natural antibody response to SARS-CoV-2 infection compared with the two groups. Lastly, we assessed the effect of dexamethasone treatment, even if statistically not significant (P > 0.05) we observed a positive trend among patients under dexamethasone in the aspect of decreasing inflammatory markers. To conclude we showed that SARS-CoV-2 is characterized by an age-dependent deregulation of inflammatory markers that are associated with mortality among COVID-19 patients.
Collapse
Affiliation(s)
- Anteneh Mehari Tizazu
- Department of Microbiology, Immunology and Parasitology, School of Medicine, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia.
| | - Addisu Gize
- Department of Microbiology, Immunology and Parasitology, School of Medicine, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
- CIHLMU Center for International Health, LMU University Hospital, LMU Munich, Germany
| | - Solomon Ali
- Department of Microbiology, Immunology and Parasitology, School of Medicine, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| |
Collapse
|
49
|
Fatoke B, Hui AL, Saqib M, Vashisth M, Aremu SO, Aremu DO, Aremu DB. Type 2 diabetes mellitus as a predictor of severe outcomes in COVID-19 - a systematic review and meta-analyses. BMC Infect Dis 2025; 25:719. [PMID: 40389865 PMCID: PMC12090609 DOI: 10.1186/s12879-025-11089-w] [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: 11/26/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic has posed significant challenges to global health, with type 2 diabetes mellitus (T2DM) emerging as a key risk factor for adverse outcomes. This study systematically reviews and quantifies the association between T2DM and COVID-19 outcomes, including mortality, severity, and need for mechanical ventilation. METHODS A systematic review and meta-analysis were conducted that adhered to PRISMA guidelines. We searched PubMed, Scopus, Web of Science and Embase for studies published from december 2019 to march 2024. Eligible studies reported on the impact of T2DM on COVID-19 outcomes in the adult population. Data were extracted and analyzed using a random-effects model, and heterogeneity was assessed using the I2 statistic. Publication bias was assessed using Egger regression, Kendall's Tau, and the Fail-safe N calculation. RESULTS Eighteen studies were included in the meta-analysis for mortality, six for severity and five for mechanical ventilation. T2DM was significantly associated with higher mortality (OR = 3.66, 95% CI: 2.20-5.11, p < 0.001), higher severity (OR = 1.97, 95% CI: 1.02-2.92, p < 0.001), and higher need for mechanical ventilation (OR = 2.34, 95% CI: 1.18-3.49, p < 0.001). Heterogeneity was high for mortality (I2 = 83.83%) but low for severity and mechanical ventilation (I2 = 0%). No significant publication bias was found. CONCLUSIONS T2DM is associated with significantly worse outcomes in COVID-19 patients, including higher mortality, higher severity and a greater likelihood of needing mechanical ventilation. These findings emphasize the need for targeted interventions and management strategies for individuals with T2DM during the ongoing pandemic. Future research should focus on understanding the underlying mechanisms and exploring strategies to mitigate these risks.
Collapse
Affiliation(s)
- Babatunde Fatoke
- General Hospital Lagos, Odan, Lagos Island, Lagos State, Nigeria
- Faculty of General Medicine, Siberian State Medical University, Tomsk, 634050, Russia
| | - Amrit Lal Hui
- Department of Psychology, National Research Tomsk State University, Tomsk, 634050, Russia
| | - Muhammad Saqib
- National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
| | - Mrinal Vashisth
- Department of Psychology, National Research Tomsk State University, Tomsk, 634050, Russia
| | - Stephen Olaide Aremu
- Faculty of General Medicine, Siberian State Medical University, Tomsk, 634050, Russia.
- Global Health and Infectious Disease Control Institute, Nasarawa State University, Keffi, Nasarawa State, PMB 1022, Nigeria.
| | | | | |
Collapse
|
50
|
Lin HF, Jiang RD, Qin RX, Yao B, Zeng WT, Gao Y, Shi AM, Li JM, Liu MQ. Characterization of a SARS-CoV-2 Infection Model in Golden Hamsters with Diabetes Mellitus. Virol Sin 2025:S1995-820X(25)00059-8. [PMID: 40389095 DOI: 10.1016/j.virs.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 05/12/2025] [Indexed: 05/21/2025] Open
Abstract
Being widespread across the globe, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) keeps evolving and generating new variants and continuously poses threat to public health, especially to the population with chronic comorbidities. Diabetes mellitus is one of high-risk factors for severe outcome of coronavirus disease 2019 (COVID-19). Establishment of animal models that parallel the clinical and pathological features of COVID-19 complicated with diabetes is thus highly essential. Here, in this study, we constructed leptin receptor gene knockout hamsters with the phenotype of diabetes mellitus (db/db), and revealed that the diabetic hamsters were more susceptible to SARS-CoV-2 and its variants than wild-type hamsters. SARS-CoV-2 and its variants induced a stronger immune cytokine response in the lungs of diabetic hamsters than in wild-type hamsters. Comparative histopathology analyses also showed that infection of SARS-CoV-2 and the variants caused more severe lung tissue injury in diabetic hamsters, and may induce serious complications such as diabetic kidney disease and cardiac lesions. Our findings demonstrated that despite the decreased respiratory pathogenicity, the SARS-CoV-2 variants were still capable of impairing other organs such as kidney and heart in diabetic hamsters, suggesting that the risk of evolving SARS-CoV-2 variants to diabetic patients should never be neglected. This hamster model may help better understand the pathogenesis mechanism of severe COVID-19 in patients with diabetes. It will also aid in development and testing of effective therapeutics and prophylactic treatments against SARS-CoV-2 variants among these high-risk populations.
Collapse
Affiliation(s)
- Hao-Feng Lin
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Laboratory Clinical Base, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China
| | - Ren-Di Jiang
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Rui-Xin Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China
| | - Bing Yao
- Jinling Hospital Department Reproductive Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Wen-Tao Zeng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China
| | - Yun Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China.
| | - Ai-Min Shi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China.
| | - Jian-Min Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China.
| | - Mei-Qin Liu
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Laboratory Clinical Base, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|