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
|
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
|
3
|
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
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
|
4
|
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
|
5
|
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. 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] [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
|
6
|
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
|
7
|
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] [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
|
8
|
Yang L, Zhou X, Liu J, Yang G, Yu J, Tan W, Fang X, Li W, He J, Ma Q, Yu L, Lu Z. Liang-Ge-San attenuates virus-induced acute lung injury by targeting FXR-mediated ACE2 downregulation to modulate the formation of the cytokine storm. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156584. [PMID: 40056637 DOI: 10.1016/j.phymed.2025.156584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Traditional Chinese medicine has been recognized for its significant role in treating acute lung injury (ALI) due to its distinct therapeutic advantages. Liang-Ge-San (LGS), a formulation from the ancient "Taiping Huimin Hejiju Fang", is believed to possess beneficial effects for treating ALI. However, LGS's precise mechanisms and efficacy in addressing viral ALI remain inadequately explored. PURPOSE To evaluate LGS's therapeutic effects and underlying mechanisms in treating viral-induced ALI. METHODS The protective effects of LGS were examined in a Polyinosinic-polycytidylic acid [Poly(I:C)]-induced ALI model using real-time quantitative PCR, enzyme-linked immunosorbent assay, and histopathological analysis. A bioinformatics approach combined with network pharmacology was utilized to ascertain the key targets of LGS in viral pneumonia. The pharmacodynamic mechanisms of LGS in viral ALI were further validated through immunofluorescence, overexpression, short hairpin RNA, chromatin immunoprecipitation, and target agonist assays. RESULTS LGS administration resulted in a reduction of IL-1β, IL-6, and TNF-α levels, along with a decrease in macrophage infiltration, pulmonary damage, and pneumonedema following the Poly(I:C) challenge. Bioinformatics and network pharmacology analyses suggested that Farnesyl X receptor (FXR) and angiotensin converting enzyme 2 (ACE2) are potential therapeutic targets for LGS in viral pneumonia. Further experiments revealed that LGS suppressed the expression of FXR, ACE2, and NF-κB-p65 in Poly(I:C)-infected cells. Notably, overexpression of FXR counteracted the repressive effects of LGS, while ACE2 expression remained unchanged in FXR-knockdown RAW264.7 cells upon treatment with Poly(I:C) or LGS. Additionally, LGS inhibited the interaction between FXR and ACE2 transcriptional promoters. In vivo, LGS attenuated the Poly(I:C)-induced upregulation of FXR, ACE2, IL-1β, IL-6, and TNF-α in ALI zebrafish and mice models, effects that could be reversed by chenodeoxycholic acid (CDCA), an FXR agonist. Moreover, LGS markedly alleviated weight loss, improved survival rates, reduced lung index, diminished viral load, and inhibited lung pathological changes in H1N1-PR8-induced ALI mice. IL-1β, IL-6, TNF-α, INF-γ, FXR, ACE2, small heterodimer partner, and NF-κB-p65 levels were markedly reduced by LGS, with these effects being reversed by CDCA. CONCLUSION This investigation provides the first evidence that FXR/ACE2 signaling is pivotal in acute respiratory viral infections, while LGS demonstrates antiviral activity against viral-induced ALI. LGS inhibits ACE2 expression induced by viral infection via FXR inhibition and modulates the cytokine storm, thus alleviating viral ALI. These findings suggest that LGS may be a promising treatment strategy for treating viral ALI.
Collapse
Affiliation(s)
- Liling Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Guangli Yang
- Department of Central Laboratory, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Weifu Tan
- Department of Neonatology, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Xiaochuan Fang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Wei Li
- Department of Neonatology, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Jiayang He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, PR China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, PR China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
9
|
Wilkinson S, Wilkinson J, Grace A, Lyon D, Mellor M, Yunus T, Manning J, Dinsdale G, Berks M, Knight S, Bakerly N, Gebril A, Dark P, Herrick A, Taylor C, Dickinson M, Murray A. Imaging the microvasculature using nailfold capillaroscopy in patients with coronavirus disease-2019; A cross-sectional study. Microvasc Res 2025; 159:104796. [PMID: 39961398 DOI: 10.1016/j.mvr.2025.104796] [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: 10/10/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVES It is understood that microvascular dysfunction plays a key role in the pathogenesis of SARS-CoV-2 coronavirus disease (COVID-19). The aim of this study was to evaluate the usefulness of an automated, quantitative nailfold capillaroscopy system in identifying microvascular changes in those confirmed with or having had COVID-19. METHODS Ninety-seven participants were enrolled into this study and grouped as follows: 52 participants with acute COVID-19 (further grouped by disease severity) and 45 participants with convalescent COVID-19 (further grouped into long COVID i.e. symptoms beyond 12 weeks, and fully recovered). Nailfold capillaroscopy images were obtained from the bilateral ring fingers using a Dino-Lite CapillaryScope 200 Pro, a small USB handheld microscope. Images were assessed quantitatively using bespoke automated measurement software and the number of haemorrhages noted for each participant. RESULTS Capillaries were predominantly 'normal' in appearance with narrow capillary loops and evenly distributed, but with an increased number of haemorrhages (40 % in the convalescent group and 17 % in the acute group, p = 0.007). There was no statistically significant difference in the mean width of capillaries (20.9-21.8 μm) or vessel density (9.6-9.9 caps/mm; acute and convalescent group, respectively). CONCLUSIONS This study has demonstrated the feasibility of nailfold capillaroscopy at the critical care bedside. Capillary structure appeared normal across all groups of individuals affected by COVID-19. Although the small differences in the microvasculature in recovered patients compared to in acutely unwell patients may suggest delayed structural change due to COVID-19, these differences are unlikely to be clinically relevant. Longitudinal studies would be required to explore this in more detail.
Collapse
Affiliation(s)
- S Wilkinson
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK; Department of Rheumatology, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - J Wilkinson
- Division of Population Health, Health Services Research & Primary Care, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK
| | - A Grace
- Emergency Assessment Unit, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - D Lyon
- Emergency Assessment Unit, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - M Mellor
- Emergency Assessment Unit, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - T Yunus
- Emergency Assessment Unit, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - J Manning
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK; Department of Rheumatology, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - G Dinsdale
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK; Department of Rheumatology, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - M Berks
- Division of Informatics, Imaging & Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK
| | - S Knight
- Lydia Becker Institute, University of Manchester, Manchester M13 9WU, UK
| | - N Bakerly
- Department of Respiratory Medicine, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - A Gebril
- Emergency Assessment Unit, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - P Dark
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK
| | - A Herrick
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK; Department of Rheumatology, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - C Taylor
- Division of Informatics, Imaging & Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK
| | - M Dickinson
- Department of Physics & Astronomy and Photon Science Institute, School of Natural Sciences, University of Manchester, M13 9PL, UK
| | - A Murray
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK; Department of Rheumatology, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK.
| |
Collapse
|
10
|
Ishigaki H, Itoh Y. Translational research on pandemic virus infection using nonhuman primate models. Virology 2025; 606:110511. [PMID: 40139071 DOI: 10.1016/j.virol.2025.110511] [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: 11/03/2024] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
After the COVID-19 pandemic, nonhuman primate (NHP) models, which are necessary for the rapid development of vaccines and new medical therapies, have become important in studies on infectious diseases because of their genetic, metabolic, and immunological similarities to humans. Our group has long been using NHP models in studies on infectious diseases including H1N1 influenza pandemic and COVID-19. Despite limitations such as the limited number of animals and the husbandry requirements, NHP models have contributed to the prediction of the pathogenicity of emerging viruses and the evaluation of the efficacy of vaccines and therapeutics due to the similarity of NHP models to humans before starting clinical trials to select good candidates of vaccines and drugs. In this review, the findings obtained in NHP infectious disease models of influenza and COVID-19 are summarized to clarify the benefits of NHP models for studies on infectious diseases. We believe that this review will support future research in exploring new perspectives for the development of vaccines and therapies targeting influenza, COVID-19, and infectious diseases in future pandemics.
Collapse
Affiliation(s)
- Hirohito Ishigaki
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, 460 Setatsukinowa, Otsu, Shiga, 520-2192, Japan
| | - Yasushi Itoh
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, 460 Setatsukinowa, Otsu, Shiga, 520-2192, Japan; Central Research Laboratory, Shiga University of Medical Science, 205 Setatsukinowa, Otsu, Shiga, 520-2192, Japan.
| |
Collapse
|
11
|
David C, Verney C, Si-Tahar M, Guillon A. Evaluating the evidence for GM-CSF as a host-directed therapy in respiratory infections. Cytokine 2025; 189:156902. [PMID: 39999678 DOI: 10.1016/j.cyto.2025.156902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
Novel therapeutic approaches are needed to treat respiratory infections due to the rising antimicrobial resistance and the lack of effective antiviral therapies. A promising avenue to overcome treatment failure is to develop strategies that target the host immune response rather than the pathogen itself. Granulocyte-macrophage colony-stimulating factor (GM-CSF) plays a critical role in controlling homeostasis in lungs, alveolar macrophages being the most sensitive cells to GM-CSF signaling. In this review, we discuss the importance of GM-CSF secretion for lung homeostasis and its alteration during respiratory infections. We also present the pre-clinical evidence and clinical investigations evaluating GM-CSF-based treatments (administration or inhibition) as a therapeutic strategy for treating respiratory infections, highlighting both supporting and contradictory findings.
Collapse
Affiliation(s)
- Camille David
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France
| | - Charles Verney
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France
| | - Antoine Guillon
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France; Université de Tours, Tours, France; CHRU de Tours, Service de Médecine Intensive Réanimation, Tours, France.
| |
Collapse
|
12
|
Fong A, Adams KT, Khairat S, Galarraga JE. Using machine learning to predict emergency department return across two regional health systems; a generalizable model for COVID-19 patients. Am J Emerg Med 2025; 91:173-174. [PMID: 39580309 DOI: 10.1016/j.ajem.2024.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024] Open
Affiliation(s)
- Allan Fong
- MedStar Health Research Institute - MedStar Health, 3007 Tilden St. NW, Suite 6N, Washington DC 20008, USA.
| | - Katharine T Adams
- MedStar Health Research Institute - MedStar Health, 3007 Tilden St. NW, Suite 6N, Washington DC 20008, USA
| | - Saif Khairat
- University of North Carolina at Chapel Hill, 428 Carrington Hall, Campus Box 7460, Chapel Hill, NC 27599, USA
| | - Jessica E Galarraga
- MedStar Health Research Institute - MedStar Health, 3007 Tilden St. NW, Suite 6N, Washington DC 20008, USA; Georgetown University School of Medicine, 3900 Reservoir Rd NW, Washington, DC 20007, USA
| |
Collapse
|
13
|
Sarnaglia AJQ, de Godoi LG, Monroy NAJ, Molinares FAF, Zamprogno B, Dias DRC, Rodrigues AS. Immunization thresholds to change the overall level and the effect of cases on deaths by COVID-19 in pregnant and postpartum women. ENVIRONMENTAL RESEARCH 2025; 271:121047. [PMID: 39947378 DOI: 10.1016/j.envres.2025.121047] [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: 07/29/2024] [Revised: 10/18/2024] [Accepted: 02/04/2025] [Indexed: 02/16/2025]
Abstract
Since the first officially reported case of COVID-19, the scientific community has spent much of its time understanding the dynamics of the virus. Several studies have indicated that some population segments are especially susceptible to COVID-19 complications, including pregnant and postpartum women. Although recommendations such as social distancing, proper sanitation, and the use of protection masks were crucial in slowing down the virus dissemination, the protection provided by vaccination is undeniable, especially for this particular group. Concerning deaths by COVID-19, it is natural to assume that daily deaths are related to reported hospitalized cases and to expect that, as vaccination increases, this effect gradually decreases. As far as we know, no other studies have addressed this issue. Therefore, this study introduces a novel generalized linear model with segmented interaction to fill this gap. The model was used to estimate the vaccination thresholds required to change the overall level and the daily hospitalized cases effect on daily deaths from COVID-19 in pregnant and postpartum women reported between January 3rd, 2021, and January 1st, 2022. Inference methods for the proposed model were developed. The results obtained indicate that, in the first period from May 25th to July 1st, 2021 (between 14,420 and 271,570 first doses, respectively), vaccination caused a significant gradual decrease in the effect of reported hospitalized cases on fatalities and, in a second period from July 25th to October 13th, 2021 (between 653,150 and 968,880 first doses, respectively), it induced a gradual reduction of the overall level of deaths. Using the average number of cases as a reference, during the period of observations, the expected number of deaths reduced from 6.16 to 0.36, a decrease of 94.16%. The importance of learning from COVID-19 data must be highlighted, as it provides us with critical insights to better prepare for future health crises.
Collapse
Affiliation(s)
- Alessandro José Queiroz Sarnaglia
- LECON, Department of Statistics, UFES, Vitória, ES, Brazil; DaSLab, Department of Statistics, UFES, Vitória, ES, Brazil.
| | - Luciana Graziela de Godoi
- LECON, Department of Statistics, UFES, Vitória, ES, Brazil; DaSLab, Department of Statistics, UFES, Vitória, ES, Brazil
| | - Nátaly Adriana Jiménez Monroy
- LECON, Department of Statistics, UFES, Vitória, ES, Brazil; DaSLab, Department of Statistics, UFES, Vitória, ES, Brazil
| | - Fabio Alexander Fajardo Molinares
- LECON, Department of Statistics, UFES, Vitória, ES, Brazil; DaSLab, Department of Statistics, UFES, Vitória, ES, Brazil
| | - Bartolomeu Zamprogno
- LECON, Department of Statistics, UFES, Vitória, ES, Brazil; DaSLab, Department of Statistics, UFES, Vitória, ES, Brazil
| | - Diego Roberto Colombo Dias
- LECON, Department of Statistics, UFES, Vitória, ES, Brazil; DaSLab, Department of Statistics, UFES, Vitória, ES, Brazil; PPGI, Department of Informatic, UFES, Vitória, ES, Brazil
| | - Agatha Sacramento Rodrigues
- LECON, Department of Statistics, UFES, Vitória, ES, Brazil; DaSLab, Department of Statistics, UFES, Vitória, ES, Brazil; PPGI, Department of Informatic, UFES, Vitória, ES, Brazil
| |
Collapse
|
14
|
Gu L, Wang ZJ, Zhang XR, Liu Y, Zhao M, Jiang SZ, Pan J, Yuan Y, Cai H, Zhou T, Li T, Li TT, Xue W. Targeting the liquid-liquid phase separation of nucleocapsid broadly inhibits the replication of SARS-CoV-2 strains. Biochem Biophys Res Commun 2025; 756:151594. [PMID: 40086356 DOI: 10.1016/j.bbrc.2025.151594] [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: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
The COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to a global public health crisis. The nucleocapsid (N) protein plays a pivotal role in a variety of biological processes in the life cycle of SARS-CoV-2, such as viral assembly. In this study, we investigated the liquid-liquid phase separation (LLPS) capacity of the N protein of seven SARS-CoV-2 strains, including the variants of concern (VOC) and interest (VOI), and its impact on viral replication. Using bioinformatic tools, we analyzed 11,433,558 complete genomes of SARS-CoV-2 and revealed a high degree of sequence conservation of N gene. While all the seven N proteins could undergo LLPS with RNA, the mutations in N impair its capacity of LLPS. With a SARS-CoV-2 trans-complementation system, we showed that SARS-CoV-2 variants carrying mutated N proteins exhibit impaired replication, highlighting the importance of LLPS of N in viral replication. We further demonstrated that (-)-gallocatechin gallate (GCG) efficiently inhibits the LLPS of N proteins and significantly suppresses the replication of different SARS-CoV-2 strains. Thus, our findings indicate that targeting the N-LLPS could be a viable strategy for the development of antiviral treatments against various SARS-CoV-2 strains, including those yet to emerge.
Collapse
Affiliation(s)
- Lin Gu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Zheng-Jie Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Xin-Ran Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Yu Liu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Ming Zhao
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Shao-Zhen Jiang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jie Pan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Ying Yuan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hong Cai
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Tao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Ting-Ting Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China.
| | - Wen Xue
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China.
| |
Collapse
|
15
|
Yang OO. The immunopathogenesis of SARS-CoV-2 infection: Overview of lessons learned in the first 5 years. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf033. [PMID: 40180332 DOI: 10.1093/jimmun/vkaf033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/11/2025] [Indexed: 04/05/2025]
Abstract
This review provides a broad overview of lessons learned in the five years since COVID-19 was identified. It is a bimodal disease, starting with an initially virus-driven phase, followed by resolution or ensuing inappropriate immune activation causing severe inflammation that is no longer strictly virus dependent. Humoral immunity is beneficial for preventing or attenuating the early stage, without benefit once the later stage begins. Neutralizing antibodies elicited by natural infection or vaccination are short-lived and highly vulnerable to viral sequence variation. By contrast, cellular immunity, particularly the CD8+ T cell arm, has a role in preventing or attenuating severe disease, is far less susceptible to viral variation, and is longer-lived than antibodies. Finally, an ill-defined phenomenon of prolonged symptoms after acute infection, termed "long COVID," is poorly understood but may involve various immunologic defects that are hyperactivating or immunosuppressive. Remaining issues include needing to better understand the immune dysregulation of severe disease to allow more tailored therapeutic interventions, developing antibody strategies that cope with the viral spike sequence variability, prolonging vaccine efficacy, and unraveling the mechanisms of long COVID to design therapeutic approaches.
Collapse
Affiliation(s)
- Otto O Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
16
|
Dhawan M, Thakur N, Sharma M, Rabaan AA. The comprehensive insights into the B-cells-mediated immune response against COVID-19 infection amid the ongoing evolution of SARS-CoV-2. Biomed Pharmacother 2025; 185:117936. [PMID: 40056829 DOI: 10.1016/j.biopha.2025.117936] [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: 09/16/2024] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025] Open
Abstract
The antibody-mediated immune response is crucial for the development of protective immunity against SARS-CoV-2, the virus responsible for the COVID-19 pandemic. Understanding the interaction between SARS-CoV-2 and the immune system is critical because new variants emerge as a result of the virus's ongoing evolution. Understanding the function of B cells in the SARS-CoV-2 infection process is critical for developing effective and long-lasting vaccines against this virus. Triggered by the innate immune response, B cells transform into memory B cells (MBCs). It is fascinating to observe how MBCs provide enduring immune defence, not only eradicating the infection but also safeguarding against future reinfection. If there is a lack of B cell activation or if the B cells are not functioning properly, it can lead to a serious manifestation of the disease and make immunisation less effective. Individuals with disruptions in the B cells have shown increased production of cytokines and chemokines, resulting in a poor prognosis for the disease. Therefore, we have developed an updated review article to gain insight into the involvement of B cells in SARS-CoV-2 infection. The discussion has covered the generation, functioning, and dynamics of neutralising antibodies (nAbs). Furthermore, we have emphasised immunotherapeutics that rely on nAbs.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, Punjab 141004, India; Trafford College, Altrincham, Altrincham, Manchester WA14 5PQ, UK.
| | - Nanamika Thakur
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Ali A Rabaan
- Research Center, Dr. Sulaiman Alhabib Medical Group, Riyadh 13328, Saudi Arabia; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan.
| |
Collapse
|
17
|
Tomos I, Antonogiannaki EM, Dimakopoulou K, Raptakis T, Apollonatou V, Kallieri M, Argentos S, Lampadakis S, Blizou M, Krouskos A, Karakatsani A, Manali E, Loukides S, Papiris S. The prognostic role of lung ultrasound in hospitalised patients with COVID-19. Correlation with chest CT findings and clinical markers of severity. Expert Rev Respir Med 2025; 19:363-370. [PMID: 40007128 DOI: 10.1080/17476348.2025.2471776] [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: 10/20/2024] [Revised: 02/08/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The use of lung ultrasound (LUS) has recently become vital in the diagnosis and prognosis of various respiratory diseases. Its role in COVID-19 requires further investigation. RESEARCH DESIGN AND METHODS Twenty-five consecutive, non-ICU hospitalized COVID-19 patients were included. LUS was performed on admission and sequentially every 3 days at 8 points in the chest. Based on the LUS findings a score was designed. Logarithmic regression models and ROC curve analysis were applied. RESULTS A statistically significant positive correlation was found between LUS score at admission and the severity of SARS-COV-2 infection. Higher LUS score was significantly associated with lower PaO2/FiO2 ratio, use of HFNC, longer hospitalization and greater extent of chest CT infiltrates. A significant association between LUS score and risk of death or intubation or HFNC was found. For one point of increase in the score, risk of death or intubation or HFNC increased 1.93-fold (95% CI 1.02 to 3.65). The predictive role of the score was very satisfactory (area under the ROC curve = 0.87). CONCLUSIONS Lung ultrasound findings were significantly positively associated with clinical and radiological markers of severity of SARS-CoV-2 pneumonia. It therefore constitutes a promising and reliable technique for assessing pneumonia, comparable to chest CT.
Collapse
Affiliation(s)
- Ioannis Tomos
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Elvira Markela Antonogiannaki
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantina Dimakopoulou
- Department of Hygiene, Epidemiology and Medical Statistics, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Thomas Raptakis
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Apollonatou
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Kallieri
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stylianos Argentos
- 2nd Department of Radiology, ATTIKON University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefanos Lampadakis
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Myrto Blizou
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonis Krouskos
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Karakatsani
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Effrosyni Manali
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stylianos Loukides
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Spyros Papiris
- 2nd Pulmonary Medicine Department, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
18
|
Song Y, Lu J, Qin P, Chen H, Chen L. Interferon-I modulation and natural products: Unraveling mechanisms and therapeutic potential in severe COVID-19. Cytokine Growth Factor Rev 2025; 82:18-30. [PMID: 39261232 DOI: 10.1016/j.cytogfr.2024.08.005] [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: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to pose a significant global public health threat, particularly to older adults, pregnant women, and individuals with underlying chronic conditions. Dysregulated immune responses to SARS-CoV-2 infection are believed to contribute to the progression of COVID-19 in severe cases. Previous studies indicates that a deficiency in type I interferon (IFN-I) immunity accounts for approximately 15 %-20 % of patients with severe pneumonia caused by COVID-19, highlighting the potential therapeutic importance of modulating IFN-I signals. Natural products and their derivatives, due to their structural diversity and novel scaffolds, play a crucial role in drug discovery. Some of these natural products targeting IFN-I have demonstrated applications in infectious diseases and inflammatory conditions. However, the immunomodulatory potential of IFN-I in critical COVID-19 pneumonia and the natural compounds regulating the related signal pathway remain not fully understood. In this review, we offer a comprehensive assessment of the association between IFN-I and severe COVID-19, exploring its mechanisms and integrating information on natural compounds effective for IFN-I regulation. Focusing on the primary targets of IFN-I, we also summarize the regulatory mechanisms of natural products, their impact on IFNs, and their therapeutic roles in viral infections. Collectively, by synthesizing these findings, our goal is to provide a valuable reference for future research and to inspire innovative treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Yuheng Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Henan University, Kaifeng 475001, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 200032, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
19
|
Tsuzuki S, Koizumi R, Asai Y, Ohmagari N. Trends in antimicrobial consumption: long-term impact of the COVID-19 pandemic. Clin Microbiol Infect 2025; 31:594-599. [PMID: 39653183 DOI: 10.1016/j.cmi.2024.12.005] [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: 07/09/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVES The COVID-19 pandemic was associated with a global decrease in antimicrobial consumption (AMC) in 2020. However, the persistence of this downwards trend is not known at a global level. This study examined the global and longer-term trends in AMC after the emergence of COVID-19. METHODS The change rate of AMC was compared (a) 2020 over 2019, (b) 2021 over 2020, and (c) 2022 over 2021 using monthly sales volume data of antimicrobials in 69 countries obtained from the IQVIA MIDAS information service. Changepoints were detected using time-series data of global monthly antimicrobial sales from November 2016 to December 2023. We defined antimicrobials as oral and parenteral drugs classified as J1 by the Anatomical Therapeutic Chemical code. Antimicrobial sales were reported in standard units, as defined by IQVIA. We assessed the data using standard units per 1000 population per day, with populations based on World Population Prospects data issued by the United Nations. In addition, interrupted time-series analysis was used to examine the impact of movement restrictions in G7 countries. RESULTS In the IQVIA MIDAS data, 68 of the 69 countries had more than one changepoint between 2016 and 2023. Of these 68 countries, 61 experienced a decrease in AMC after the COVID-19 pandemic started. However, 53 of these 61 countries showed a reverse increasing trend in AMC in 2022. Interrupted time-series analysis revealed that movement restrictions had a negative impact on AMC in all G7 countries. DISCUSSION The global decrease in AMC in 2020 might not have been because of COVID-19 itself but to non-pharmaceutical interventions such as movement restrictions. Human mobility could possibly be one of the key determinants of antimicrobial use at the population level.
Collapse
Affiliation(s)
- Shinya Tsuzuki
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan; Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.
| | - Ryuji Koizumi
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yusuke Asai
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan; Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan; Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Barrachina-Esteve O, Anguita A, Reverter A, Espinosa J, Lafuente C, Rubio-Roy M, Crosas M, Vila-Sala C, Acero C, Navarro M, Cánovas D, Ribera G, Jodar M, Estela J. Neurologic features in hospitalized patients with COVID-19: a prospective cohort in a catalan hospital. Neurol Sci 2025; 46:1477-1488. [PMID: 39951175 PMCID: PMC11920300 DOI: 10.1007/s10072-025-08031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVES To study the prevalence and timing of neurological manifestations, including cognitive involvement, in patients hospitalized for Coronavirus disease 2019 (COVID-19). To analyze the pathogenic mechanisms and any association they have with disease severity. METHODS Longitudinal cohort study with prospective follow-up of patients who required hospitalization. Patients under 65 who had no pre-existing cognitive impairment and did not require an ICU stay were evaluated 3 and 12 months after discharge using a battery of neuropsychological tests. RESULTS Of 205 patients hospitalized for COVID-19, 153 (74.6%) presented with neurological manifestations. The most frequent were myalgia (32.7%), headache (31.7%), dysgeusia (29.2%), and anosmia (24.9%). Patients with more severe illness at the time of hospitalization presented fewer neurological manifestations. Of the 62 patients who underwent neuropsychological examination 3 months after discharge, 22.6% had impaired attention, 19.4% impaired working memory, 16.1% impaired learning and retrieval, 9.7% impaired executive functions, and 8.2% impaired processing speed. Patients with anosmia also presented with more headache (OR 5.45; p < 0.001) and greater risk of working memory impairment (OR 5.87; p 0.03). At follow-up 12 months after hospital discharge, 14.3% of patients still showed impaired attention, 2.4% impaired working memory, 2.5% impaired executive functions, and 2.5% impaired processing speed. DISCUSSION Neurological manifestations are common in patients hospitalized for COVID-19 regardless of severity. The high prevalence of anosmia and its association with headache and working memory impairment at 3 months, suggest potential direct or indirect damage to the prefrontal cortex via invasion of the olfactory bulb by COVID-19.
Collapse
Affiliation(s)
- Oriol Barrachina-Esteve
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain.
- Department of Neurology, Manacor Hospital, Manacor, Mallorca, Spain.
| | - A Anguita
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
- Department of Neurology, Sant Joan de Déu Hospital, Althaia Foundation, Manresa, Spain
| | - A Reverter
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - J Espinosa
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Lafuente
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Rubio-Roy
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Crosas
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Vila-Sala
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Acero
- Department of Ophthalmology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Navarro
- Department of Infectious Diseases, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - D Cánovas
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - G Ribera
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Jodar
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
- Department of Clinical and Health Psychology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), National Institute of Health Carlos III, Madrid, Spain
| | - J Estela
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| |
Collapse
|
21
|
Zhao Y, Xue Y, Wang C, Zhao Z, Cui R, Zhu B. Antibacterial poly(ethyl methacrylate) surfaces constructed by facile amination with polyethyleneimine of different architectures. Colloids Surf B Biointerfaces 2025; 248:114458. [PMID: 39724827 DOI: 10.1016/j.colsurfb.2024.114458] [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: 10/26/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
Polymethacrylate and its derivatives are widely used in food industry and biomedical applications for their plasticity, biocompatibility and optical transparency. However, susceptibility to bacterial growth on their surfaces limits their applications. In this study, linear and branched polyethyleneimine (PEI) molecules were grafted onto poly(ethyl methacrylate) (PEMA) via aminolysis using a simple one-step method to enhance the antibacterial properties of PEMA films. PEI-modified PEMA films were characterized by ATR-FTIR, X-ray photoelectron spectroscopy (XPS), field emission scanning electron microscopy (FESEM) and thermal gravimetric analysis (TGA). The modified films exhibited optimal bactericidal efficiency of 98.0 % against Escherichia coli (E. coli) and over 99.9 % against Staphylococcus aureus (S. aureus). Furthermore, hydrolysis was found to contribute to anchoring PEI onto PEMA as well. Though branched PEI exhibited a higher grafting amount than the linear ones under same conditions, PEMA modified with linear PEI presented a similar or even higher antibacterial efficiency than those grafted with branched PEI. Overall, PEI-grafted PEMA films prepared with simple one-step method exhibit effective antibacterial properties and good biocompatibilities, making them promising candidates for biomedical devices and other applications.
Collapse
Affiliation(s)
- Yu Zhao
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yunyun Xue
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center of Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China
| | - Chuyao Wang
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zihao Zhao
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ronglu Cui
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Baoku Zhu
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center of Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China.
| |
Collapse
|
22
|
Kanda R, Miyazaki Y, Nakayamada S, Fukuyo S, Kubo S, Miyagawa I, Yamaguchi A, Satoh-Kanda Y, Ohkubo N, Todoroki Y, Tanaka H, Ueno M, Nagayasu A, Fujita Y, Aritomi T, Kusaka K, Sakai H, Matsunaga S, Nohara H, Tanaka Y. Effective Second-Line b/tsDMARDs for Patients with Rheumatoid Arthritis Unresponsive to First-Line b/tsDMARDs from the FIRST Registry. Rheumatol Ther 2025; 12:353-369. [PMID: 40025347 PMCID: PMC11920512 DOI: 10.1007/s40744-025-00747-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/23/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION For patients with rheumatoid arthritis (RA) unresponsive to first-line biologic/targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs), the selection of second-line b/tsDMARDs is crucial to prevent progression to difficult-to-treat rheumatoid arthritis (D2TRA). However, indicators for selection are lacking. This study aimed to identify optimal second-line b/tsDMARDs among the phase III treatment strategies based on European League Against Rheumatism (EULAR) RA management recommendations. METHODS A total of 687 RA patients treated with second-line b/tsDMARDs (tumor necrosis factor inhibitor (n = 246), interleukin-6 receptor inhibitor [n = 195], cytotoxic T-lymphocyte-associated protein 4 immunoglobulin [n = 119], and Janus kinase inhibitor [n = 127]) were enrolled between October 2013 and April 2023. Rates of patients achieving Clinical Disease Activity Index (CDAI) remission and CDAI low disease activity (LDA), changes in CDAI, persistence rates, and adverse events within 24 weeks after treatment initiation were compared among the four groups. Propensity score-based inverse probability of treatment weighting (PS-IPTW) was used to minimize selection bias. RESULTS After PS-IPTW adjustment, the Janus kinase inhibitor (JAKi) group had the highest persistence rate among the four groups. At 24 weeks, the JAKi group showed the greatest improvement in CDAI and the highest CDAI remission rate. Among patients treated with JAKi as second-line b/tsDMARDs, upadacitinib (UPA) was most likely to achieve CDAI remission at 24 weeks. The comparison between the UPA group (n = 32) and the non-UPA JAKi group (tofacitinib and baricitinib [n = 95]) showed comparable persistence rates but significantly lower CDAI scores and higher CDAI remission rate at 24 weeks in the UPA group. No significant difference was noted in the overall incidence of adverse events among the four groups treated with b/tsDMARDs or between the groups treated with JAKi. CONCLUSIONS Selecting JAKi, especially UPA, may effectively improve the disease activity for RA patients unresponsive to first-line b/tsDMARDs. Further large-scale studies are needed to clarify the efficacy and safety of UPA. TRIAL REGISTRATION FIRST registry (approval number#04-23): October 2013, retrospectively registered.
Collapse
Affiliation(s)
- Ryuichiro Kanda
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yusuke Miyazaki
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Shingo Nakayamada
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Shunsuke Fukuyo
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Satoshi Kubo
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Molecular Targeted Therapies, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Ippei Miyagawa
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Ayako Yamaguchi
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Laboratory and Transfusion Medicine, Hospital of the University of the Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yurie Satoh-Kanda
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Naoaki Ohkubo
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Environmental Epidemiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yasuyuki Todoroki
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Molecular Targeted Therapies, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Hiroaki Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Masanobu Ueno
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Atsushi Nagayasu
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yuya Fujita
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Takafumi Aritomi
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Laboratory and Transfusion Medicine, Hospital of the University of the Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Katsuhide Kusaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Hidenori Sakai
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Satsuki Matsunaga
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Hirotsugu Nohara
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan.
| |
Collapse
|
23
|
Hasani H, Hamidi F, Ahmadi-Forg F, Panahi P, Tofighi Khelejan F. The Effect of Prior Use of Statins on the Severity of COVID-19 Disease: A Retrospective Study. Crit Care Nurs Q 2025; 48:143-150. [PMID: 40009860 DOI: 10.1097/cnq.0000000000000544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
It has been suggested that the use of statin pills beforehand could potentially influence the outcomes when individuals are hospitalized with COVID-19. In this study, we investigated how the prior use of statin medication could influence the COVID-19 severity parameters. In this retrospective cohort study, we categorized COVID-19 patients into 2 groups: statin users and non-users. Then, various data including age, gender, the patient's need for ventilation support, the lowest oxygen blood saturation level, the length of hospitalization, receiving remdesivir treatment, and their COVID-19 vaccination status were collected. Out of 168 patients, 62 had taken statin medication before being admitted. Using statins decreased the patient's need for ventilation support, length of hospitalization, ventilation duration, and oxygen saturation level (P < .001). Interaction effect analysis showed that receiving remdesivir statically affected the length of hospitalization, ventilation duration, and oxygen saturation level but did not significantly affect the association between statins and needing to ventilator. The use of statin pills before COVID-19 admission reduced the requirement for ventilator support.
Collapse
Affiliation(s)
- Hadi Hasani
- Author Affiliations: Department of Nursing, School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran (Mr Hasani); Department of Statistics and Epidemiology, Faculty of Health, Tabriz University of Medical Sciences, Tabriz, Iran (Ms Hamidi); Department of Nursing, Tabas School of Nursing, Birjand University of Medical Sciences, Birjand, Iran (Ms Ahmadi-Forg); Student research committee, School of Nursing and Midwifery, Islamic Azad University of Dezful, Dezful, Iran (Ms Panahi); and Department of Mathematics and Statistics, Faculty of Science, Dalhousie university, Nova Scotia, Canada (Dr Tofighi Khelejan)
| | | | | | | | | |
Collapse
|
24
|
Soni DK, Cabrera-Luque J, Kar S, Ahmed A, Sen C, Devaney J, Biswas R. Suppression of miR-155 Attenuates Lung Cytokine Storm Induced by SARS-CoV-2 Infection. J Interferon Cytokine Res 2025; 45:150-161. [PMID: 39950973 DOI: 10.1089/jir.2024.0253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a deadly human viral disease with a high rate of infection, morbidity, and mortality. Although vaccines and antiviral treatments are available, hospitalizations remain steady, and concerns about long-term consequences persist. Therefore, there is a great urgency to develop novel therapies. Here, we analyzed the role of miR-155, one of the most powerful drivers of host antiviral responses including immune and inflammatory responses, in the pathogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Endogenous microRNAs (miRNAs, miRs) are key molecules in preventing viral entry and replication while building an antiviral cellular defense. Our study reveals that miR-155 expression is elevated in patients with COVID-19. Using a mouse model transgenic for human angiotensin-converting enzyme receptor 2, we evaluated the potential of anti-miR-155 therapy. Treating SARS-CoV-2-infected mice with anti-miR-155 significantly reduced miR-155 expression, improved survival, and slightly increased body weight. Notably, these mice showed altered expression of cytokines in the lungs. These findings suggest anti-miR-155 could be a promising therapy to mitigate the cytokine storm and long-lasting symptoms induced by SARS-CoV-2 infection, improving public health outcomes and enhancing global pandemic preparedness.
Collapse
Affiliation(s)
- Dharmendra Kumar Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | | | - Anwar Ahmed
- Department of Preventive Medicine and Biostatistics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Chaitali Sen
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
25
|
Mukherjee S, Bayry J. The Yin and Yang of TLR4 in COVID-19. Cytokine Growth Factor Rev 2025; 82:70-85. [PMID: 39490235 DOI: 10.1016/j.cytogfr.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 11/05/2024]
Abstract
Various pattern recognition receptors (PRRs), including toll-like receptors (TLRs), play a crucial role in recognizing invading pathogens as well as damage-associated molecular patterns (DAMPs) released in response to infection. The resulting signaling cascades initiate appropriate immune responses to eliminate these pathogens. Current evidence suggests that SARS-CoV-2-driven activation of TLR4, whether through direct recognition of the spike glycoprotein (alone or in combination with endotoxin) or by sensing various TLR4-activating DAMPs or alarmins released during viral infection, acts as a critical mediator of antiviral immunity. However, TLR4 exerts a dual role in COVID-19, demonstrating both beneficial and deleterious effects. Dysregulated TLR4 signaling is implicated in the proinflammatory consequences linked to the immunopathogenesis of COVID-19. Additionally, TLR4 polymorphisms contribute to severity of the disease. Given its significant immunoregulatory impact on COVID-19 immunopathology and host immunity, TLR4 has emerged as a key target for developing inhibitors and immunotherapeutic strategies to mitigate the adverse effects associated with SARS-CoV-2 and related infections. Furthermore, TLR4 agonists are also being explored as adjuvants to enhance immune responses to SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory (IBIL), Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal 713 340, India.
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris 75006, France; Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678 623, India.
| |
Collapse
|
26
|
Tcholadze G, Pantsulaia I, Ratiani L, Kopaleishvili L, Bolotashvili T, Jorbenadze A, Chikovani T. The Prognostic Value of Circulating Cytokines and Complete Blood Count-Based Inflammatory Markers in COVID-19 Patients With Atrial Fibrillation. Cardiol Res 2025; 16:153-160. [PMID: 40051670 PMCID: PMC11882233 DOI: 10.14740/cr2027] [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: 12/06/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Background Atrial fibrillation (AF) is associated with a high burden of cardiovascular disease, which has been worsened during the coronavirus disease 2019 (COVID-19) pandemic. The purpose of this study was to assess the association between clinical markers, especially interleukin-6 (IL-6) and other inflammatory biomarkers, and the severity of COVID-19 in patients with AF. Methods This retrospective cohort study categorized patients based on clinical presentations and laboratory results to investigate the prognostic significance of inflammatory markers in COVID-19 outcomes among those with AF. The study included 100 hospitalized COVID-19 patients aged between 40 to 80 years and was conducted at the Chapidze Hospital in Tbilisi, Georgia. Patients were then grouped by disease severity according to computed tomography (CT) scores, clinical symptoms, respiratory rate and oxygen saturation. Levels of IL-6 were obtained at three time points during hospitalization. A broad range of laboratory tests, including C-reactive protein (CRP), ferritin, and D-dimer, were also conducted. Results Patients with AF demonstrated significantly elevated levels of IL-6 (P = 0.024), CRP (P = 0.001), and ferritin (P < 0.001), suggesting a severe inflammatory response. D-dimer levels were also notably higher in the AF group (P < 0.005), indicating an increased risk of thrombotic complications. Oxygen saturation levels were significantly lower (P = 0.004) and CT scores higher in patients with AF. Furthermore, the length of hospitalization was longer among patients with AF (median duration significantly higher, P = 0.032), indicating a more severe disease course. Conclusions The proinflammatory markers such as IL-6 are independent predictive markers of COVID-19 severity in AF patients. Overall, it highlights urgent treatment approaches, such as available anti-inflammatory drugs, for COVID-19 patients with arrhythmias. Combining these biomarkers into clinical routines helps us better identify patients at risk and how to treat them.
Collapse
Affiliation(s)
- Giorgi Tcholadze
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| | - Ia Pantsulaia
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
- Vl. Bakhutashvili Institute of Medical Biotechnology, Tbilisi State Medical University, Tbilisi 0159, Georgia
| | | | | | | | | | - Tinatin Chikovani
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| |
Collapse
|
27
|
Nouanesengsy A, Semesi A, Quach K, Ivanochko D, Byrne W, Hwang M, La Neve MR, Leon-Ponte M, Litosh A, Wisener N, Adeli K, Campigotto A, Grunebaum E, McGeer A, Moraes TJ, Sepiashvili L, Upton J, Julien JP, Allen U. Persistence and decay of neutralizing antibody responses elicited by SARS-CoV-2 infection and hybrid immunity in a Canadian cohort. Microbiol Spectr 2025; 13:e0133324. [PMID: 39969224 PMCID: PMC11960127 DOI: 10.1128/spectrum.01333-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
A major challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been assessing the intensity, dynamics, and determinants of the antibody responses after infection and/or vaccination. Therefore, we aimed to characterize the longitudinal dynamics of the antibody responses among naturally infected individuals and individuals who achieved hybrid immunity in a large Canadian cohort. We demonstrate that anti-Spike IgGs and neutralizing antibody dynamics vary greatly among individuals with COVID-19, in peak antibody levels, rate of waning, and longevity of the antibody response. Additionally, we found an association between robust antibody responses and individuals with severe COVID-19 clinical symptoms during the first-month post-symptom onset. For individuals who achieved hybrid immunity, a robust increase in anti-S1 IgGs and neutralizing antibodies followed the first vaccination dose; however, there was a minimal increase in the anti-S1 IgGs and neutralizing antibody titers after administration of the second dose of the vaccine. Furthermore, neutralizing antibodies elicited by the wild-type virus alone were largely ineffective against emerging variants of concern in our natural infection-only cohort, in contrast to a much broader and more robust neutralization profile observed in individuals who achieved hybrid immunity. Our findings emphasize the need for global SARS-CoV-2 vaccination efforts to further sustain protective immune responses required to minimize viral spread and disease severity in the population. As SARS-CoV-2 variants continue to emerge, understanding the interplay between previous infections, vaccine durability, and virus evolution will be critical for guiding ongoing vaccination strategies. IMPORTANCE A major challenge with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been assessing the intensity, dynamics, and determinants of the antibody response after infection and/or vaccination. Our paper addresses this in a large Canadian cohort with antibody responses that were generated by natural infection as well as vaccine in some persons studied.
Collapse
Affiliation(s)
- Amy Nouanesengsy
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Anthony Semesi
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Kim Quach
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Walter Byrne
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matthew Hwang
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maria-Rosa La Neve
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matilde Leon-Ponte
- Division of Allergy and Immunology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alice Litosh
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicole Wisener
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Khosrow Adeli
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Aaron Campigotto
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Eyal Grunebaum
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
| | - Allison McGeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tunenbaum Research Institute at Mount Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Theo J. Moraes
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lusia Sepiashvili
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Upton
- Division of Allergy and Immunology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Upton Allen
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, The Hospital for Sick Children, Research Institute, Toronto, Ontario, Canada
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Victorin D, Bergquist H, Hafsten L, Nihlén Å, Lindell E. Low Molecular Weight Heparin Dosing in Relation to Postoperative Bleeding After Tracheotomy in Patients Infected With SARS-CoV-2-A Descriptive Study. Laryngoscope Investig Otolaryngol 2025; 10:e70122. [PMID: 40177253 PMCID: PMC11963079 DOI: 10.1002/lio2.70122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/02/2025] [Accepted: 02/02/2025] [Indexed: 04/05/2025] Open
Abstract
Objective The aim of this study was to analyze whether patients with SARS-CoV-2 who received surgical tracheotomy had a lower incidence of postoperative bleeding if their LMWH was postponed or canceled on the day of surgery. Methods Patients with SARS-CoV-2 admitted to the intensive care units who underwent surgical tracheotomy were assessed retrospectively through their medical records. Data on comorbidity, LMWH dose, and timing were collected. Bleedings < 72 h post surgery were noted as stomal or airway bleedings. Results All 101 patients included were on LMWH. Twenty-two patients had no change of dose of LMWH, 24 patients had their dose of LMWH postponed to post surgery, and 50 patients had their dose reduced to only the evening dose on the day of surgery. Twenty-six patients had a stomal bleeding, one patient had an airway bleeding, and four patients had both stomal and airway bleedings. No significant difference in the incidence of bleeding was identified between various groups of different LMWH doses or timing, reduced dose versus no change of dose, OR 1.29 (95% CI 0.42-3.92). Postponed dose versus no change of dose of LMWH, OR 1.03 (95% CI 0.28-3.75). Increasing age was correlated to a higher risk of bleeding post-surgery by an OR of 1.64 (95% CI 1.06-2.54, p = 0.026 for every 10 years added). No fatal bleeding related to surgical tracheotomy was observed. Conclusion Decreased doses of LMWH on the day of surgery were not associated with a risk reduction for post-surgical bleeding in patients with SARS-CoV-2 who received tracheotomy. Increasing age was a risk factor for post-surgical bleeding. Level of Evidence Retrospective, level 3.
Collapse
Affiliation(s)
- David Victorin
- Department of OtorhinolaryngologyRegion Västra Götaland, Södra Älvsborg HospitalBoråsSweden
- Department of Research, Education and InnovationRegion Västra Götaland, Södra Älvsborg HospitalBoråsSweden
| | - Henrik Bergquist
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of OtorhinolaryngologyRegion Västra Götaland, Sahlgrenska University HospitalGothenburgSweden
| | - Louise Hafsten
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Åsa Nihlén
- Department of OtorhinolaryngologyRegion Västra Götaland, Södra Älvsborg HospitalBoråsSweden
| | - Ellen Lindell
- Department of OtorhinolaryngologyRegion Västra Götaland, Södra Älvsborg HospitalBoråsSweden
- Department of Otorhinolaryngology, Head and Neck Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
29
|
Zhang L, Li F, Liu X, Liu XA, Lu D, Luo Q, Liu Q, Jiang G. Long-term effects of SARS-CoV-2 infection on metal homeostasis. J Trace Elem Med Biol 2025; 88:127625. [PMID: 40023939 DOI: 10.1016/j.jtemb.2025.127625] [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: 11/21/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
The outbreak of COVID-19 pandemic has caused substantial health loss worldwide, and the long-term sequelae of COVID, resulting from repeated coronavirus infection, have emerged as a new public health concern. We report the widespread presence of abnormal metallomic profiles in the sera of patients who have recovered from SARS-CoV-2 coronavirus infection, even after 6 months post-discharge from hospital. We measured the concentrations of Fe, Cu, Zn, Se, Cr, Mn, Ba, Ni, Pb, Ag, As, Cd, Co, and V in the sera of 25 recovered participants and 38 healthy controls in the cross-sectional study. Higher concentrations of Cu, Ag, As, Ba, Cd, Ni, Pb, Cr and V were observed in the recovered participants, whereas lower concentrations of Fe and Se were obtained in these participants. Except for Zn, Mn, and Co, all other elements showed significant differences (p < 0.05) between the two groups, with variations dependent on age and gender. Further correlation analysis between metallome and metabolome indicated that SARS-CoV-2 infection continues to disrupt metallic homeostasis and affect metabolic processes, such as lipid metabolism and cell respiration, as well as the functions of certain organs (e.g., liver, kidney, and heart), even after 6 months recovery. Our findings provide novel insights into the potential long-term effect of COVID-19 on the human body from a new perspective of metallomics.
Collapse
Affiliation(s)
- Luyao Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Fang Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Xiaoxiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Xin-An Liu
- Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Dawei Lu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qian Luo
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Qian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
30
|
Schwarze M, Brakel A, Hoffmann R, Krizsan A. Peptides Corresponding to the Receptor-Binding Domain (RBD) of Several SARS-CoV-2 Variants Of Concern Prevent Recognition of the Human ACE2 Receptor and Consecutive Cell Infections. ChemMedChem 2025; 20:e202400973. [PMID: 39996354 DOI: 10.1002/cmdc.202400973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Indexed: 02/26/2025]
Abstract
New strategies are needed to prevent and control upcoming outbreaks of SARS-CoV-2 infections, independent of vaccination. SARS-CoV-2 binds to the human ACE-2 receptor through the receptor binding domain (RBD) of the spike (S) protein, allowing the virus to enter human cells and begin replication. When peptides corresponding to four regions of RBD containing previously reported ACE-2 interaction sites were explored, the sequence 392 to 421, peptide p392wt, bound strongly to ACE-2 and inhibited wild-type RBD binding to ACE-2. Interestingly, p392 peptides corresponding to mutated sequences from different SARS-CoV-2 VOCs, including the current VOC BA.5 and KP.3, bound less strongly to ACE-2, but showed partially better inhibition of the ACE-2 interaction of all tested RBDs. When studied in a SARS-CoV-2 pseudovirus assay, the p392 peptides showed a good inhibition rate of 98.8±8.1 % at a peptide concentration of ~244 μmol/L, while none of the p392 peptides inhibited antibody binding to the RBD, suggesting that peptide treatment is sufficient in the presence of anti-RBD antibodies. Interestingly these peptides were active in the presence of diluted human serum and non-toxic to human cell lines.
Collapse
Affiliation(s)
- Mandy Schwarze
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Alexandra Brakel
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Ralf Hoffmann
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Andor Krizsan
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, Leipzig, Germany
- Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
31
|
Spector BL, Koseva B, McLennan R, Banerjee D, Lankachandra K, Bradley T, Selvarangan R, Grundberg E. Methylation patterns of the nasal epigenome of hospitalized SARS-CoV-2 positive patients reveal insights into molecular mechanisms of COVID-19. BMC Med Genomics 2025; 18:62. [PMID: 40170038 PMCID: PMC11963311 DOI: 10.1186/s12920-025-02125-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/12/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has varied presentations from asymptomatic to death. Efforts to identify factors responsible for differential COVID-19 severity include but are not limited to genome wide association studies (GWAS) and transcriptomic analysis. More recently, variability in host epigenomic profiles have garnered attention, providing links to disease severity. However, whole epigenome analysis of the respiratory tract, the target tissue of SARS-CoV-2, remains ill-defined. RESULTS We interrogated the nasal methylome to identify pathophysiologic drivers in COVID-19 severity through whole genome bisulfite sequencing (WGBS) of nasal samples from COVID-19 positive individuals with severe and mild presentation of disease. We noted differential DNA methylation in intergenic regions and low methylated regions (LMRs), demonstrating the importance of distal regulatory elements in gene regulation in COVID-19 illness. Additionally, we demonstrated differential methylation of pathways implicated in immune cell recruitment and function, and the inflammatory response. We found significant hypermethylation of the FUT4 promoter implicating impaired neutrophil adhesion in severe disease. We also identified hypermethylation of ELF5 binding sites suggesting downregulation of ELF5 targets in the nasal cavity as a factor in COVID-19 phenotypic variability. CONCLUSIONS This study demonstrated DNA methylation as a marker of the immune response to SARS-CoV-2 infection, with enhancer-like elements playing significant roles. It is difficult to discern whether this differential methylation is a predisposing factor to severe COVID-19, or if methylation differences occur in response to disease severity. These differences in the nasal methylome may contribute to disease severity, or conversely, the nasal immune system may respond to severe infection through differential immune cell recruitment and immune function, and through differential regulation of the inflammatory response.
Collapse
Affiliation(s)
- Benjamin L Spector
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI, 53792, USA.
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA.
| | - Boryana Koseva
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Rebecca McLennan
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Dithi Banerjee
- Department of Pathology and Laboratory Medicine, Children'S Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Kamani Lankachandra
- Department of Pathology, University Health, University of Missouri- Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Todd Bradley
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Rangaraj Selvarangan
- Department of Pathology and Laboratory Medicine, Children'S Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Elin Grundberg
- Department of Pediatrics, Genomic Medicine Center, Children's Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO, 64108, USA.
| |
Collapse
|
32
|
Fang XM, Liu XJ, Zhang RG. Carbon monoxide inhibits human bronchial epithelial CCL5 and IL-6 secretion induced by SARS-CoV-2 spike RBD protein. Exp Cell Res 2025; 447:114499. [PMID: 40058446 DOI: 10.1016/j.yexcr.2025.114499] [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: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Carbon monoxide (CO) is a novel anti-inflammatory molecule, but the effects of CO on SARS-CoV-2 spike RBD (S-RBD)-induced human bronchial epithelial cytokines release remains unclear. CO was delivered using CO-releasing molecule 3 (CORM-3). The effects of S-RBD, ATPγS and CO on cytokines secretion were determined by enzyme-linked immunosorbent assay (ELISA) in 16HBE14o-human bronchial epithelial cell line. The inhibitory effect of CO on S-RBD-induced ERK phosphorylation was assessed by Western blot analysis. The regulatory effect of CO on extracellular nucleotide-induced ion transport was quantified by short-circuit current (ISC). S-RBD evoked CCL5 and IL-6 release and this effect could be suppressed by CO. However, CO failed to inhibit ATP release induced by S-RBD while decreased ATP-induced CCL5 and IL-6 secretion as well as ion transport. Furthermore, CO significantly inhibited ERK phosphorylation induced by S-RBD. These findings suggest an anti-inflammatory role of CO during inflammation induced by S-RBD and extracellular nucleotide in human bronchiol epithelial cells.
Collapse
Affiliation(s)
- Xiao-Min Fang
- Department of Physiology, Basic Medical School, Guangdong Medical University, Zhanjiang, China
| | - Xing-Jian Liu
- Department of Physiology, Basic Medical School, Guangdong Medical University, Zhanjiang, China
| | - Rui-Gang Zhang
- Department of Physiology, Basic Medical School, Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
33
|
Lai C, Lu S, Yang Y, You X, Xu F, Deng X, Lan L, Guo Y, Kuang Z, Luo Y, Yuan L, Meng L, Wu X, Song Z, Jiang N. Myeloid-Driven Immune Suppression Subverts Neutralizing Antibodies and T Cell Immunity in Severe COVID-19. J Med Virol 2025; 97:e70335. [PMID: 40183283 PMCID: PMC11969634 DOI: 10.1002/jmv.70335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025]
Abstract
The objective of this study was to better understand immune failure mechanisms during severe acute respiratory syndrome coronavirus 2, SARS-CoV-2 infection, which are critical for developing targeted vaccines and effective treatments. We collected 34 cases representing different disease severities and performed high-quality single-cell TCR/BCR sequencing to analyze the peripheral immune cell profiles. Additionally, we assessed antibody-neutralizing activity through in vitro experiments. Our integrated multiomics analysis uncovers a profound immune paradox in severe COVID-19: hyperinflammation coexists with immunosuppression, driven by distinct yet interconnected dysregulatory mechanisms. Severe patients develop robust humoral immunity, evidenced by clonally expanded plasma cells producing neutralizing antibodies (e.g., IGHG1-dominated responses) and antigen-specific T cell activation. However, these protective responses are counteracted by myeloid-driven immunosuppression, particularly CD14+ HMGB2+ monocytes exhibiting metabolic reprogramming and HLA-DR downregulation, coupled with progressive T cell exhaustion characterized by IFN-γ/TNF-α hyperactivation and impaired antigen presentation. Importantly, prolonged viral persistence in severe cases arises from a failure to coordinate humoral and cellular immunity-antibody-mediated neutralization cannot compensate for defective cytotoxic T cell function and monocyte-mediated immune suppression. These findings highlight the necessity for therapeutic strategies that simultaneously enhance antibody effector functions (e.g., Fc optimization), restore exhausted T cells, and reverse myeloid suppression. They also highlight the importance of vaccines designed to elicit balanced B cell memory and durable T cell responses, which are critical to preventing severe disease progression. By addressing the dual challenges of hyperinflammation and immunosuppression, such approaches could restore immune coordination and improve outcomes in severe COVID-19.
Collapse
Grants
- This work was supported by the National Key Research and Development Program of China (2021YFC2501800, 2022YFA0806200, 2023YFC0872500, and 2024YFC3044600), the National Natural Science Foundation of China (82072214, 82272198, and 82202373), the Science and Technology of Shanghai Committee (21MC1930400, 22Y11900100, and 23Y31900100), and the Shanghai Municipal Health Commission (2023ZDFC0101).
Collapse
Affiliation(s)
- Cong Lai
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Su Lu
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yilin Yang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Xiaoyu You
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Feixiang Xu
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Xinran Deng
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Lulu Lan
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yuesheng Guo
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Zhongshu Kuang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yue Luo
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Li Yuan
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Lu Meng
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Xueling Wu
- Department of Respiratory MedicineShanghai Jiaotong University School of Medicine, Renji HospitalShanghaiChina
| | - Zhenju Song
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Ning Jiang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| |
Collapse
|
34
|
Wang L, Mi R, Chen L, Liu J, Yang H, Hu M, Xiaoqiang Z, Zhang Y, Xu X, Liu B, Zhao H, Qianyu L, Liu T, Zhenzhu C, Yao J, Yang Y, Wei X. Clinical Characteristics of SARS-COV-2 Omicron Variant in Acute Myeloid Leukemia and Acute Lymphocytic Leukemia Patients: A Multi-Center Retrospective Study. Cancer Rep (Hoboken) 2025; 8:e70146. [PMID: 40176607 PMCID: PMC11965881 DOI: 10.1002/cnr2.70146] [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: 03/24/2024] [Revised: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND The death rate of hematological malignancies is high, and the death rate of patients with COVID-19 infection is further increased. Although there have been expert consensus and relevant guidelines to introduce the recommendations of the guidelines for patients with hematological malignancies complicated with COVID-19 infection, there is limited understanding of the clinical characteristics of Chinese patients with acute leukemia complicated with COVID-19 infection. AIMS This study aimed to analyze the clinical manifestations, mortality, and determinants of viral shedding duration in Chinese AL patients infected with COVID-19. METHODS We conducted a retrospective study of 100 AL patients with COVID-19 infection in Henan Province, China, from December 1, 2022, to January 31, 2023. Data on demographics, leukemia subtype, symptoms, treatments (antibiotics/antivirals), and viral shedding duration were collected. Follow-up was conducted over three months to assess mortality. Univariate and multivariate analyses were performed to identify risk factors. RESULTS The median age was 49.5 years (58% male, 42% female), with 76% having acute myeloid leukemia (AML) and 24% acute lymphoblastic leukemia (ALL). Most patients (86%) were asymptomatic. Antibiotics and antivirals were administered to 35% and 25% of patients, respectively. Severe cases and fatalities exhibited prolonged viral shedding. Neutropenic patients on antibiotics had significantly extended shedding duration, whereas antiviral therapy or delayed primary disease management shortened it. The overall mortality rate was 6%. Univariate analysis identified neutropenia as a key mortality risk factor, though multivariate analysis showed no significant associations. CONCLUSION Early antiviral treatment may reduce viral shedding duration and potentially mitigate symptom severity and mortality in AL patients with COVID-19. Neutropenia emerged as a critical factor influencing outcomes. These findings underscore the importance of tailored therapeutic strategies for this high-risk population.
Collapse
Affiliation(s)
- Lin Wang
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer HospitalZhengzhouChina
| | - Ruihua Mi
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer HospitalZhengzhouChina
| | - Lin Chen
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer HospitalZhengzhouChina
| | - Jia Liu
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer HospitalZhengzhouChina
| | - Haiping Yang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and TechnologyLuoyangChina
| | - Meng Hu
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and TechnologyLuoyangChina
| | - Zhao Xiaoqiang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and TechnologyLuoyangChina
| | | | | | - Bing Liu
- Pingdingshan First People HospitalPingdingshanChina
| | | | - Li Qianyu
- Huaihe Hospital of Henan UniversityKaifengChina
| | - Tao Liu
- Zhoukou Central HospitalZhoukouChina
| | | | - Jinxiao Yao
- Nanyang Second People's HospitalNanyangChina
| | - Ying Yang
- Nanyang Second People's HospitalNanyangChina
| | - Xudong Wei
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer HospitalZhengzhouChina
| |
Collapse
|
35
|
Fraisse A, Guillier L, Cordevant C, Le Poder S, Perelle S, Martin-Latil S. Impedance-based method for the quantification of infectious SARS-CoV-2. J Virol Methods 2025; 333:115110. [PMID: 39855472 DOI: 10.1016/j.jviromet.2025.115110] [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: 08/12/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiologic agent involved in the coronavirus disease 2019 (COVID-19) pandemic. The development of infectious titration methods is crucial to provide data for a better understanding of transmission routes, as well as to validate the efficacy of inactivation treatments. Nevertheless, the low-throughput analytical capacity of traditional methods may be a limiting factor for a large screening of samples. The aim of the study was to develop a Real-Time Cell Analysis (RTCA) assay based on the measurement of cell impedance to quantify infectious SARS-CoV-2. The kinetics of cell impedance showed a virus-specific Cell Index (CI) drop. This enabled the correlation between viral concentrations and time at which a 50 % drop in CI values was observed (tCI50), with establishment of a standard curve. In parallel, the improved Spearman and Kärber method was used to quantify infectious titer since the virus-induced CI drop is correlated to the Cytopathic Effect. The estimated uncertainty was respectively 0.57, 0.36, and 0.26 log10 with 4, 8, and 16 wells per dilution. Thus, the RTCA assay is a powerful tool with a greatly simplified workflow for effective risk assessment in the field of food and environmental virology.
Collapse
Affiliation(s)
- Audrey Fraisse
- Université Paris-Est, ANSES, Laboratory for food safety, Maisons-Alfort F-94700, France
| | - Laurent Guillier
- ANSES, Risk Assessment Department, Maisons-Alfort F-94700, France
| | - Christophe Cordevant
- ANSES, Strategy and Programs Department, Research and Reference Division, Maisons-Alfort F-94 700, France
| | - Sophie Le Poder
- UMR VIROLOGIE, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort F-94700, France
| | - Sylvie Perelle
- Université Paris-Est, ANSES, Laboratory for food safety, Maisons-Alfort F-94700, France
| | - Sandra Martin-Latil
- Université Paris-Est, ANSES, Laboratory for food safety, Maisons-Alfort F-94700, France; UMR VIROLOGIE, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort F-94700, France.
| |
Collapse
|
36
|
Winyupakorn J, Sangketchon C, Devakul Na Ayutthaya W, Sethasine S. Liver injury in non-severe COVID-19 with various pandemic phases: A real-world study. J Formos Med Assoc 2025:S0929-6646(25)00144-5. [PMID: 40169313 DOI: 10.1016/j.jfma.2025.03.026] [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: 02/27/2024] [Revised: 10/15/2024] [Accepted: 03/26/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND COVID-19 severity affects liver damage. The utilization of various anti-COVID-19 drugs in non-severe cases related to liver impairment in the short term seemed intriguing. OBJECTIVES To assess the dynamic course of liver injury in mild to moderate COVID-19 patients within 10 days of admission and identify risk variables, including medication linkage. METHODS This prospective cohort study of 300 newly diagnosed mild to moderate COVID-19 cases between September 2021 and October 2022. Tertiary center hospitel, field hospital, or cohort ward admissions were made. Patient demographics and treatment were recorded. The drug, liver injury (LI) dynamics, and clinical course were evaluated. RESULTS Hospitel/field hospital (188) and cohort wards (112) had 300 individuals. One hundred fifteen participants had liver damage. Favipiravir (45 %), remdesivir (17.4 %), molnupiravir (11.3 %), Andrographis paniculata (ADG) (8.7 %), and favipiravir plus ivermectin (7.7 %) were given to most LI group (n = 104). The baseline AST/ALT levels of 68 (65.4 %) treated patients were abnormal. Favipiravir, remdesivir, and favipiravir + ivermectin increased mean AST/ALT in participants with normal baseline AST/ALT (p = 0.001, 0.003, and 0.016, respectively), but not molnupiravir or Andrographis paniculata. Transaminase levels climbed in untreated patients independent of baseline. The ground-glass imaging pattern was linked to mild LI. Most subjects had transaminase declines after 10 days. Preexisting liver disease did not increase the likelihood of in-hospital LI. CONCLUSION In the real world, a less-than-critical level of liver damage was reported in mild to moderate COVID-19 that allows clinicians to administer a variety of standard medications during short periods of hospital stay.
Collapse
Affiliation(s)
- Jirayuth Winyupakorn
- Division of Gastroenterology and Hepatology, Department of Medicine, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, 10300, Thailand
| | - Chunlanee Sangketchon
- Division of Disaster and Emergency, Medical Operation Department, Faculty of Science and Health Technology, Vajira Hospital, Navamindradhiraj University, Dusit, 10300, Bangkok, Thailand
| | - Watcharaporn Devakul Na Ayutthaya
- Division of Pharmacology, Department of Basic Medical Science, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, 10300, Thailand
| | - Supatsri Sethasine
- Division of Gastroenterology and Hepatology, Department of Medicine, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, 10300, Thailand.
| |
Collapse
|
37
|
Reagin KL, Oliva KE, Hansen MR, Slade CD, Watford WT, Klonowski KD. Regulation of respiratory CD8+ T-cell immunity by suppressive monocyte-like dendritic cells (MCs). JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae059. [PMID: 40163680 DOI: 10.1093/jimmun/vkae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/12/2024] [Indexed: 04/02/2025]
Abstract
Active immune suppression can mediate the balance between protective cellular immunity and harmful immunopathology. This suppression can occur locally, at an infection site, or in regional draining lymph nodes (dLNs). Immune regulation is of particular importance in sites such as the lung where aberrant immunopathology can result in loss of tissue function and respiratory failure. We have recently identified a novel population of CD11b+CD103+CCR2+ monocyte-like dendritic cells (MCs) which directly suppress CD8+ T-cell proliferation in vitro. Respiratory infection of mice with RNA viruses recruits these MCs either exclusively to the dLN (after vesicular stomatitis virus infection) or both the dLN and site of viral replication (after influenza infection). Here we show that depletion of MCs from the dLN of mice using CCR2-DTR bone marrow chimeras results in enhanced respiratory CD8+ T-cell responses and lung tissue-resident memory cell (TRM) formation which correlated with enhanced antiviral responses upon heterologous VSV challenge. Conversely, depletion of MCs from both the dLN and respiratory tract following influenza infection results in enhanced respiratory CD8+ T-cell responses coupled with fatal immunopathology. Together, these data suggest that suppressive MCs govern key aspects of respiratory CD8+ T-cell immunity, thereby balancing immunity and adverse pathology in the context of viral infection.
Collapse
Affiliation(s)
- Katie L Reagin
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Kimberly E Oliva
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Matthew R Hansen
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Chris D Slade
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Wendy T Watford
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Kimberly D Klonowski
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| |
Collapse
|
38
|
Zhuang Q, Zhu J, Peng B, Zhu Y, Cheng K, Ming Y. Correlation between peripheral lymphocyte subsets monitoring and COVID-19 pneumonia in kidney transplant recipients. BMC Infect Dis 2025; 25:426. [PMID: 40148763 PMCID: PMC11948920 DOI: 10.1186/s12879-025-10581-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/30/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVES In kidney transplant recipients (KTRs), immune monitoring of peripheral lymphocyte subsets (PLS) reflects the real immune status and aids in the diagnosis of the occurrence and development of infectious diseases, including COVID-19. Exploring the PLS of COVID-19 pneumonia in KTRs is important. METHODS In this study, a total of 103 KTRs were divided into mild pneumonia (MP) and severe pneumonia (SP) groups, as well as a stable group. The clinical information and PLS data were assessed via t or Mann-Whitney test and receiver operating curve analysis. Logistic regression was employed to identify the risk factors, and Spearman's correlation analysis was used to identify correlations. RESULTS Lymphopenia is a common manifestation of COVID-19 in KTRs, and it is positively related to the severity of COVID-19 pneumonia. The CD3 + T-cell count had the highest AUC between the MP and the SP. Multiple PLS measures were found to be independent risk factors for COVID-19 pneumonia progression in KTRs. CONCLUSIONS This study revealed a robust correlation between PLS and severe COVID-19 pneumonia progression in KTRs. PLS monitoring could facilitate real-time diagnosis and therapy for infection, as well as timely and precisive adjustment of immunosuppression instructions, for KTRs with COVID-19.
Collapse
Affiliation(s)
- Quan Zhuang
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Jiang Zhu
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Bo Peng
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Yi Zhu
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Ke Cheng
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Yingzi Ming
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China.
| |
Collapse
|
39
|
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 2025:107004. [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] [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
|
40
|
Phillips CM, Smyth JW. Viral Infection and Connexin Dysfunction in the Heart. Curr Cardiol Rep 2025; 27:76. [PMID: 40146392 PMCID: PMC11950093 DOI: 10.1007/s11886-025-02227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 03/28/2025]
Abstract
PURPOSE OF REVIEW Gap junctions, comprising connexin proteins, enable the direct intercellular electrical coupling of cardiomyocytes, and disruption of this process is arrhythmogenic. In addition, gap junctions effect metabolic coupling and of relevance to this review, propagate host antiviral immune responses. Accordingly, connexins have emerged as viral targets during infection. This review summarizes current knowledge regarding contributions of inflammation vs virally encoded factors in driving alterations to cardiac gap junction function. RECENT FINDINGS In addition to host immune-mediated effects on cardiac electrophysiology and gap junctions in myocarditis, there is now increasing appreciation for virally encoded factors targeting connexin function in acute/active infection. We now know diverse viral species have independently evolved to directly target connexin function during infection. Understanding both the direct and indirect effects of viral infection on cardiac gap junctions is critical to inform treatment strategies and development of novel therapeutics for acute infection as a distinct disease process from chronic myocarditis.
Collapse
Affiliation(s)
- Chelsea M Phillips
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
| | - James W Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA.
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
41
|
He R, Zhang J, Tian Y, Yan J, Huang J, Sun T, Xie Y, Pu W, Wu T. Integrating multiplex PCR in fever clinics for acute respiratory pathogen-specific diagnosis. Clin Chim Acta 2025; 572:120245. [PMID: 40157701 DOI: 10.1016/j.cca.2025.120245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
The epidemiological patterns of respiratory tract infections (RTIs) have experienced substantial changes due to the influence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with a particular focus on acute respiratory infections (ARIs). Challenges in early diagnosis, inadequate triage strategies, and the inappropriate use of antimicrobials or antivirals have compounded the difficulties in accurately diagnosing and managing ARIs in the post-pandemic context. This study aimed to investigate the efficacy of fever clinics equipped with nucleic acid testing capabilities in the precise triage of ARIs. In a cohort of 604 individuals presenting with symptoms of ARIs, we utilized real-time reverse transcription polymerase chain reaction (RT-PCR) technology available in the fever clinic to perform nucleic acid testing for SARS-CoV-2, influenza A virus (Flu A), influenza B virus (Flu B), respiratory syncytial virus, adenovirus, human rhinovirus, and Mycoplasma pneumoniae. Subsequently, statistical methods were employed to analyze the distribution and types of ARIs associated with these pathogens. In fever clinics, most patients presenting with respiratory pathogen infections were diagnosed with non-SARS-CoV-2 respiratory pathogens, with a higher incidence noted among pediatric patients compared to adults. In contrast, SARS-CoV-2 primarily affected the adult population and was linked to more severe clinical outcomes. Consequently, the swift triage of patients exhibiting ARI symptoms in a fever clinic equipped with nucleic acid testing enables the rapid identification and precise treatment of pathogens. This approach alleviates patient discomfort and enhances the efficiency of healthcare resource utilization.
Collapse
Affiliation(s)
- Ruifen He
- Department of Clinical Laboratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China
| | - Jianwen Zhang
- Department of Clinical Laboratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China
| | - Yuan Tian
- Public Health Center, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China
| | - Junxia Yan
- Department of Clinical Laboratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China
| | - Jinjuan Huang
- Department of Clinical Laboratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China
| | - Tingting Sun
- Department of Clinical Laboratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China
| | - Yuxin Xie
- Department of Clinical Laboratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China
| | - Wenjia Pu
- Department of Clinical Laboratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China
| | - Tao Wu
- Department of Clinical Laboratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan 750001, China.
| |
Collapse
|
42
|
Awundja BMA, Tchuente BRT, Hagbe VP, Nnanga LS, Kamdem L, Ekedjoum DYE, Roger OMH, Maguerrite B, Patricia D, Ndanji JA, Mbah LJN, Lumngwena EN, Bongue B, Ngondi JL. Biological profile and risk factors of mortality in COVID-19 patients at Adlucem hospital in Banka-Bafang, Cameroon: a cross-sectional study. BMC Infect Dis 2025; 25:420. [PMID: 40141010 PMCID: PMC11948750 DOI: 10.1186/s12879-025-10845-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/21/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Despite availability of preventive vaccine and global control of the SARS-CoV-19 transmission, continuous emergence of new strains coupled with the increase spread of Mpox poses significant public health threats. Identification of simple factors for stratification and prognostics of hospitalized patients is crucial for management of these patients in limited resource settings. The aim of this study was to assess the biological profile of severe hospitalized COVID-19 patients in Cameroon and identify risk factors for mortality. METHODS A prospective, cross-sectional, analytical study was conducted of a cohort of COVID-19 patients admitted and managed at the Adlucem hospital in Banka-Bafang, Haut-Nkam Department, West Cameroon Region, from 2021 to 2022. The clinical characteristics and biological parameters of patients with COVID-19 were evaluated. RESULTS Of the 259 cases of COVID-19 included in the study, 68 cases (26.3%) died. The majority of patients who died were over 70 years of age. Key factors predictive of mortality in these patients were leukocytosis (OR = 2.035; 95%CI: 1.161-3.567; p = 0.013), thrombocytosis (OR = 4.286; 95%CI: 1.152-15.950; p = 0.030), hypokalemia (OR = 2.400; 95%CI: 1.143-5.042; p = 0.021), hyponatremia (OR = 2.292; 95%CI: 1.185-4.431; p = 0.014) and hypochloremia (OR = 2.644; 95%CI: 1.188-5.882; p = 0.017). CONCLUSION Age, electrolyte imbalance and thrombocytosis were predictive of death in COVID-19 patients in this cohort. Thus, a biological work-up should be considered for risk stratification to ensure efficient management of COVID-19 patients on a case-by-case basis in resource limited settings like Cameroon. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
| | - Boris Ronald Tonou Tchuente
- Department of Biochemistry, Faculty of Sciences, University of Yaounde 1, Yaounde, P.O. Box 812, Cameroon
- Centre for Food, Food Security and Nutrition Research, Institute of Medical Research and Medicinal Plant Studies, Yaounde, Cameroon
| | - Vervaine Pauline Hagbe
- Department of Biochemistry, Faculty of Sciences, University of Yaounde 1, Yaounde, P.O. Box 812, Cameroon
| | - Leila Sandra Nnanga
- Department of Biochemistry, Faculty of Sciences, University of Yaounde 1, Yaounde, P.O. Box 812, Cameroon
- Centre for Research on Medicinal Plants and Traditional Medicine, Institute of Medical Research and Medicinal Plant Studies, Yaoundé, Cameroon
| | - Linda Kamdem
- INSERM, U1059, DVH Team, SAINBIOSE, Jean Monnet University Saint-Etienne, University Hospital of Saint-Etienne, Saint-Etienne, France
| | | | | | | | | | | | - Leonel Javeres Ntepe Mbah
- Laboratory of Human Metabolism and Non-Communicable Disease, Institute of Medical Research and Medicinal Plant Studies, Yaounde, Cameroon
| | - Evelyn Ngwa Lumngwena
- School of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
- Centre for the study of Emerging and re-Emerging pathogens (CREMER), Institute for Medical Research and Medicinal Plant Studies, Yaoundé, Cameroun
| | - Bienvenu Bongue
- INSERM, U1059, DVH Team, SAINBIOSE, Jean Monnet University Saint-Etienne, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Judith Laure Ngondi
- Department of Biochemistry, Faculty of Sciences, University of Yaounde 1, Yaounde, P.O. Box 812, Cameroon.
- Centre of Nutrition and Functional Foods, PO Box 8024, Yaoundé, Cameroon.
| |
Collapse
|
43
|
Steinberg R, Troxler S, Dac LH, Kentgens AC, Bovermann X, Aebi C, Frey U, Bittel P, Agyeman P, Latzin P, Korten I. Changes in respiratory viruses in infancy during the SARS-CoV-2 pandemic: a prospective cohort study. BMJ Open Respir Res 2025; 12:e003044. [PMID: 40139841 PMCID: PMC11950960 DOI: 10.1136/bmjresp-2024-003044] [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: 11/22/2024] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Respiratory virus infections are a major cause of morbidity in early life. During the SARS-CoV-2 pandemic, non-pharmaceutical interventions (NPIs) lead to worldwide changes in respiratory virus epidemiology. However, evidence regarding virus circulation in the outpatient setting remains largely unknown. The aim of this study is to longitudinally assess respiratory viruses in healthy infants before and during the SARS-CoV-2 pandemic in Switzerland. METHODS In this prospective observational birth cohort study, we followed 34 infants throughout the first year of life before and during the SARS-CoV-2 pandemic. We analysed 648 biweekly nasal swabs for nine different respiratory viruses by Multiplex-PCR and assessed respiratory symptoms, COVID-19 infections of family members and childcare status in weekly interviews. 712 nasal swabs from 32 infants analysed before the pandemic and published previously served as control group. RESULTS During the period with strict NPIs (pandemic I), most common respiratory viruses were not detected, with a rebound (driven by Adenovirus and Parainfluenza virus) after most NPIs were relaxed (pandemic II): prepandemic: 27%, pandemic I: 19%, pandemic II: 33%; historic: 36% of collected swabs per period, p<0.001. Human rhinovirus (HRV) prevalence persisted during NPIs presence, mainly in the form of asymptomatic HRV detection: prepandemic=24%, pandemic I=19%, pandemic II=25%, historic: 25%, p=0.3. SARS-CoV-2 detection (asymptomatic and symptomatic) was low, and only present after NPIs were relaxed: pandemic II=2.4%. No severe COVID-19 infections were reported. DISCUSSION In our cohort, infants did not contribute largely to spread of SARS-CoV-2. The role of persisting asymptomatic HRV prevalence is still unclear, but it might help to maintain population immunity to prevent more severe infections. Our results underscore the importance of capturing asymptomatic viruses via longitudinal community-based data assessment to better understand virus transmission.
Collapse
Affiliation(s)
- Ruth Steinberg
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Simone Troxler
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Léa Ho Dac
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Anne-Christianne Kentgens
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xenia Bovermann
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Christoph Aebi
- Division of Paediatric Infectious Diseases, Department of Paediatrics, University of Bern, Bern, Switzerland
| | - Urs Frey
- Pediatrics, Basel University Children's Hospital (UKBB), Basel, Switzerland
| | - Pascal Bittel
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Philipp Agyeman
- Division of Paediatric Infectious Diseases, Department of Paediatrics, University of Bern, Bern, Switzerland
| | - Philipp Latzin
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Insa Korten
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
44
|
Ali KA, He LX, Gao F, Xia ZA, Huang H, Zeng H, Hu WH. Pathogen Detection in Spinal Infections: Next-Generation Sequencing Versus Conventional Microbiological Methods. Curr Med Sci 2025:10.1007/s11596-025-00040-4. [PMID: 40138143 DOI: 10.1007/s11596-025-00040-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE AND BACKGROUND Early and accurate diagnosis of spinal infections, including spinal tuberculosis, is pivotal for effective treatment but remains challenging. This study aims to assess the diagnostic yield of metagenomic next-generation sequencing (mNGS) compared with that of conventional microbiological tests (CMTs) in identifying pathogens associated with spinal pathologies, with a special focus on infections leading to surgical interventions. METHODS We enrolled 85 patients who underwent spinal surgery, comprising 63 patients with clinically diagnosed spinal infections, including patients with spinal tuberculosis, and 22 patients with noninfectious spinal conditions. The procedures involved irrigation and debridement for persistent wound drainage, with subsequent DNA extraction from plasma and joint fluid for mNGS and CMT analysis. RESULTS Significantly increased C-reactive protein (CRP) levels were observed in patients with infections. The mNGS approach showed greater diagnostic sensitivity (92.06%) for detecting pathogens, including Mycobacterium tuberculosis, than did CMTs (36.51%). Despite its low specificity, mNGS had considerable negative predictive value (70.59%), underscoring its utility in ruling out infections. CONCLUSIONS The mNGS offers superior sensitivity over CMTs in the diagnosis of a variety of spinal infections, notably spinal tuberculosis. This study highlights the potential of mNGS in enhancing the diagnosis of complex spinal infections, thereby informing targeted treatment strategies.
Collapse
Affiliation(s)
- Khan Akhtar Ali
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling-Xiao He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fang Gao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ze-An Xia
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Heng Zeng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wei-Hua Hu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
45
|
Sánchez A, García-Pardo G, Martí A, Gómez-Bertomeu F, Chafino S, Massanella M, Flores-Piñas M, Cedó L, Vidal F, Peraire J, Rull A. Omics for searching plasma biomarkers associated with unfavorable COVID-19 progression in hypertensive patients. Sci Rep 2025; 15:10343. [PMID: 40133696 PMCID: PMC11937446 DOI: 10.1038/s41598-025-94725-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Hypertension is one of the most common risk factors for COVID-19 clinical progression. The identification of plasma biomarkers for anticipating worse clinical outcomes and to better understand the shared mechanisms between hypertension and COVID-19 are needed. A hypothesis-generating study was designed to compare plasma proteomics and metabolomics between 22 hypertensives (HT) and 41 non-hypertensives (nHT) patients with the most unfavorable COVID-19 progression. A total of 43 molecules were significantly differed between HT (n = 22) and nHT (n = 41). Random Forest (RF) analysis identified myo-inositol, gelsolin and phosphatidylcholine (PC) 32:1 as the top molecules for distinguishing between HT and nHT. Plasma myo-inositol and gelsolin were higher (P = 0.03 and P = 0.02, respectively) and plasma PC 32:1 was lower (P = 0.03) in HT compared to nHT. Biological processes like stress response and blood coagulation, along with KEGG pathways including ascorbate and aldarate metabolism (P = 0.021) and linoleic acid metabolism (P = 0.028), were altered in hypertensive patients with the most unfavorable COVID-19 progression. There is a clear link between hypertension and severe COVID-19. Key biological pathways to consider for improving the prognosis and quality of life of hypertensive patients who become infected with SARS-CoV-2 include oxidative stress, ascorbate and aldarate metabolism, lipid metabolism, immune system and inflammation.
Collapse
Affiliation(s)
- Alba Sánchez
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Graciano García-Pardo
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Anna Martí
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Frederic Gómez-Bertomeu
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Silvia Chafino
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Massanella
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- IrsiCaixa, Hospital Universitari Germans Trias i Pujol, 08916, Badalona, Spain
| | - Marina Flores-Piñas
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
| | - Lídia Cedó
- Grup de Recerca en Diabetis i Malalties Metabòliques Associades (DIAMET), Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Joan XXIII, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Francesc Vidal
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Joaquim Peraire
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
- Universitat Rovira i Virgili (URV), Tarragona, Spain.
| | - Anna Rull
- Infection and Immunity (INIM), Institut Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII (HJ23), Tarragona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
- Universitat Rovira i Virgili (URV), Tarragona, Spain.
| |
Collapse
|
46
|
Bhanu P, Buchke S, Hemandhar-Kumar N, Varsha P, Kiran SKR, Vikneswaran G, Alva A, Basavaraj GS, Kumar J. Comparative metagenomic analysis of the oral microbiome in COVID-19 patients and healthy individuals. Sci Rep 2025; 15:10303. [PMID: 40133298 PMCID: PMC11937335 DOI: 10.1038/s41598-024-81864-3] [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: 09/19/2024] [Accepted: 11/29/2024] [Indexed: 03/27/2025] Open
Abstract
COVID-19, caused by SARS-CoV-2, affects multiple body systems, including the oral cavity, where it may disrupt the oral microbiome in ways that contribute to disease pathology. Understanding the long-term interaction between SARS-CoV-2 and the oral microbiome is crucial, as it may reveal microbial markers valuable for diagnosing or monitoring persistent health issues in COVID-19 survivors. Metagenomic sequencing revealed significant microbial shifts in the oral microbiome of COVID-19 patients, showing reduced microbial diversity and increased prevalence of opportunistic pathogens compared to healthy individuals. Alpha diversity measures indicated lower microbial diversity and evenness, while beta diversity analyses demonstrated distinct microbial community compositions. Core microbiome analysis identified unique taxa in COVID-19 patients that may contribute to disease pathology, while differential abundance analysis highlighted specific taxa shifts, including an increase in potential pathogens. Our findings advance the understanding of microbial changes in the oral microbiome associated with COVID-19 and suggest potential targets for microbiome-based interventions. While these results indicate associations with possible health impacts, further research is needed to determine causative links and long-term implications for COVID-19 survivors. This foundational research highlights the potential for microbiome science to inform diagnostic tools, such as microbial markers for disease progression, and therapeutic approaches, including targeted probiotics, which could ultimately support better patient outcomes and public health strategies.
Collapse
Affiliation(s)
- Piyush Bhanu
- Xome Life Sciences Pvt. Ltd., Bangalore Bioinnovation Centre, Helix Biotech Park, Electronic City, Bangalore, India.
- School of Engineering, Department of Bioengineering, University of Kansas, 1450 Jayhawk Blvd, Lawrence, KS, 66045, USA.
| | - Sakshi Buchke
- Xome Life Sciences Pvt. Ltd., Bangalore Bioinnovation Centre, Helix Biotech Park, Electronic City, Bangalore, India
- Morsani College of Medicine, University of South Florida, 12901 Bruce B., Downs Blvd., Tampa, FL, 33612, USA
| | - Nisha Hemandhar-Kumar
- Xome Life Sciences Pvt. Ltd., Bangalore Bioinnovation Centre, Helix Biotech Park, Electronic City, Bangalore, India
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Piyush Varsha
- Xome Life Sciences Pvt. Ltd., Bangalore Bioinnovation Centre, Helix Biotech Park, Electronic City, Bangalore, India
- Saraswati Dental College, Tiwari Ganj, 233, Faizabad Rd, Uattardhona, Uttar Pradesh, 226028, India
| | - S K Ravi Kiran
- Narayana Hrudayalaya Hospital, Narayana Hrudayalaya Ltd., Bommasandra Industrial Area, Hosur Road, Anekal Taluk, Bangalore, Karnataka, India
| | - G Vikneswaran
- Narayana Hrudayalaya Hospital, Narayana Hrudayalaya Ltd., Bommasandra Industrial Area, Hosur Road, Anekal Taluk, Bangalore, Karnataka, India
| | - Arjun Alva
- Narayana Hrudayalaya Hospital, Narayana Hrudayalaya Ltd., Bommasandra Industrial Area, Hosur Road, Anekal Taluk, Bangalore, Karnataka, India
| | - G S Basavaraj
- Narayana Hrudayalaya Hospital, Narayana Hrudayalaya Ltd., Bommasandra Industrial Area, Hosur Road, Anekal Taluk, Bangalore, Karnataka, India
| | - Jitendra Kumar
- Biotechnology Industry Research Assistance Council (BIRAC), H728+H5G, NSIC Estate, Okhla Phase III, Okhla Industrial Estate, New Delhi, Delhi, India.
| |
Collapse
|
47
|
Owolabi IJ, Karim SU, Khanal S, Valdivia S, Frenzel C, Bai F, Flynt AS. Processing of genomic RNAs by Dicer in bat cells limits SARS-CoV-2 replication. Virol J 2025; 22:86. [PMID: 40133950 PMCID: PMC11934715 DOI: 10.1186/s12985-025-02693-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Bats are reservoirs for numerous viruses that cause serious diseases in other animals and humans. Several mechanisms are proposed to contribute to the tolerance of bats to these pathogens. This study investigates the response of bat cells to double-stranded RNA generated by SARS-CoV-2 replication. Here, we found the involvement of Dicer in the processing of viral genomic RNAs during SARS-CoV-2 infection. Examining RNA sequencing of infected cells, small-interfering RNA (siRNA)-like fragments were found derived from viral RNAs. Depletion of Dicer showed a reduction in these RNAs and an increase in viral loads suggesting unlike other mammals, bats may use Dicer to limit viral replication. This prompted the exploration of key dsRNA sensors in bat cells. Our analysis showed significant upregulation of OAS1 and MX1 in response to dsRNA, while PKR levels remained low, suggesting alternative dsRNA-response mechanisms are present that eschew the common PKR-based system. These results further show how bats employ distinct strategies for antiviral defense that may contribute to tolerating viral infections. They suggest the involvement of Dicer in antiviral mechanisms in bats, a function not observed in other mammals. This highlights a mechanism for bat originating viruses to evolve features that in other animals could cause extreme antiviral responses such as is seen with SARS-CoV-2.
Collapse
Affiliation(s)
- Iyanuoluwani J Owolabi
- Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Shazeed-Ul Karim
- Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Sweta Khanal
- Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Sergio Valdivia
- Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Christopher Frenzel
- Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Fengwei Bai
- Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Alex S Flynt
- Cellular and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS, 39406, USA.
| |
Collapse
|
48
|
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] [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
|
49
|
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] [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
|
50
|
He Y, Zheng Q, Zhifang Z, Xiaofeng N, Shenggen W, Xue M, Zheng C, Liu Z. When COVID-19 meets diabetes: A bibliometric analysis. Diabetes Res Clin Pract 2025; 223:112118. [PMID: 40132732 DOI: 10.1016/j.diabres.2025.112118] [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: 11/01/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Coronavirus disease 2019 (COVID-19) survivors are concerned about the likelihood of developing further diseases. This study examines the global trends in scientific research on diabetes associated with COVID-19 from several perspectives. Bibliometric analyses are used to undertake a scientific review of the literature. The Web of Science Core Collection (WoSCC) database was used to acquire bibliographic information on diabetes related to COVID-19 from Jan 2020 to Dec. 2023. The visual map was built via advanced CiteSpace 6.2.R6. 7,348 papers were found. Khunti Kamlesh and Rizzo-Manfredi are the most well-known high-yield authors in this area, and the top ten authors collaborate extensively. Most of these papers came from universities. Harvard Medical School has the most publications, followed by Wuhan University and Huazhong University of Science and Technology. China and the United States are the countries with the most publications. Angiotensin-converting enzymes, chronic disease, intensive care unit, viral infection, and gestational diabetes mellitus were scored 0-11, 2, 3, and 4, respectively. Zhou et al.'s work on this topic, which appeared in the prominent medical journal The Lancet, was cited 1,366 times, highlighting its importance. "clinical characteristics," "diabetes mellitus," "metabolic syndrome," and "angiotensin-converting enzyme" were used as keywords for reference co-citation and clustering data identify. Over the last four years, related investigations have focused primarily on observing clinical aspects. This report is important for developing treatment strategies, directing future research, and guiding clinical practice.
Collapse
Affiliation(s)
- Yingli He
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Qingcong Zheng
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhang Zhifang
- Fujian Center for Disease Control and Prevention, Fuzhou 350012, China
| | | | - Wu Shenggen
- Fujian Center for Disease Control and Prevention, Fuzhou 350012, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Zhijun Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, China.
| |
Collapse
|