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
|
Ma L, Lin X, Xu M, Ke X, Liu D, Chen Q. Exploring the biological mechanisms of severe COVID-19 in the elderly: Insights from an aged mouse model. Virulence 2025; 16:2487671. [PMID: 40228062 PMCID: PMC12005417 DOI: 10.1080/21505594.2025.2487671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 02/04/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025] Open
Abstract
The elderly population, who have increased susceptibility to severe outcomes, have been particularly impacted by the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), leading to a global health crisis. However, definitive parameters or mechanisms underlying the severity of COVID-19 in elderly people remain unclear. Thus, this study seeks to elucidate the mechanism behind the increased vulnerability of elderly individuals to severe COVID-19. We employed an aged mouse model with a mouse-adapted SARS-CoV-2 strain to mimic the severe symptoms observed in elderly patients with COVID-19. Comprehensive analyses of the whole lung were performed using transcriptome and proteome sequencing, comparing data from aged and young mice. For transcriptome analysis, bulk RNA sequencing was conducted using an Illumina sequencing platform. Proteomic analysis was performed using mass spectrometry following protein extraction, digestion, and peptide labelling. We analysed the transcriptome and proteome profiles of young and aged mice and discovered that aged mice exhibited elevated baseline levels of inflammation and tissue damage repair. After SARS-CoV-2 infection, aged mice showed increased antiviral and inflammatory responses; however, these responses were weaker than those in young mice, with significant complement and coagulation cascade responses. In summary, our study demonstrates that the increased vulnerability of the elderly to severe COVID-19 may be attributed to an attenuated antiviral response and the overactivation of complement and coagulation cascades. Future research on antiviral and inflammatory responses is likely to yield treatments that reduce the severity of viral respiratory diseases in the elderly.
Collapse
Affiliation(s)
- Li Ma
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xian Lin
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Meng Xu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xianliang Ke
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Di Liu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Quanjiao Chen
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
5
|
Kaga M, Sasaki M, Masuda T, Sato H, Ueda T, Hirakawa A. Exploring the association between happy hypoxia and Coronavirus disease 2019 in the triage phase. Future Sci OA 2025; 11:2458413. [PMID: 39882841 PMCID: PMC11792821 DOI: 10.1080/20565623.2025.2458413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Patients with severe coronavirus disease 2019 (COVID-19) have been reported to show hypoxia without displaying typical clinical signs or symptoms, called "happy hypoxia." To explore the potential of happy hypoxia as a distinctive symptom of COVID-19, we compared vital signs in the triage phase between patients with and without COVID-19. METHODS We retrospectively identified emergency patients with and without COVID-19 admitted to Rakuwakai Marutamachi Hospital, Kyoto, Japan, between January 2021 and December 2021. RESULTS AND CONCLUSIONS 317 patients were analyzed. Multivariate logistic regression analysis, including all vital signs, demonstrated that the respiratory rate was not statistically associated with COVID-19 (odds ratio, 0.94, p = 0.058), suggesting that happy hypoxia may not be a distinct hallmark of COVID-19.
Collapse
Affiliation(s)
- Mihiro Kaga
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
- Emergency and General Internal Medicine, Rakuwakai Marutamachi Hospital, Kyoto, Japan
| | - Masanao Sasaki
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Takahiro Masuda
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
- Department of Intensive Care, Institute of Science Tokyo, Tokyo, Japan
| | - Hiroyuki Sato
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Takeshi Ueda
- Emergency and General Internal Medicine, Rakuwakai Marutamachi Hospital, Kyoto, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Pini L, Giordani J, Levi G, Guerini M, Piva S, Peli E, Violini M, Piras S, El Masri Y, Pini A, Visca D, Assanelli D, Muiesan ML, Latronico N, Tantucci C. 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
|
7
|
Kalbaugh DV. Dynamics of an epidemic controlled by isolation and quarantine: A probability-based deterministic model. Infect Dis Model 2025; 10:813-839. [PMID: 40271143 PMCID: PMC12017978 DOI: 10.1016/j.idm.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/25/2025] Open
Abstract
Assuming a homogeneous population, we employ a deterministic model based on first principles of probability to explore dynamics of an epidemic controlled by isolation alone, quarantine alone, and the two together. We develop explicit closed-form equations for key metrics of control performance: cumulative fraction of population infected over the course of the epidemic (final size), maximum fraction infected at any one time, and epidemic duration. We derive an analytical solution for final size of an epidemic controlled by isolation, when final size is small, and develop empirical relations for the other cases. We frame equations in terms of reproduction numbers, measures of intervention effort and initial conditions. We model both strength and speed of interventions, assume second order gamma distributions for intervention waiting times and employ non-time-invariant equations for quarantine. We also account for quarantine of unexposed, susceptible individuals and for imperfect intervention.
Collapse
|
8
|
Trier NH, Zivlaei N, Ostrowski SR, Sørensen E, Larsen M, Slibinskas R, Ciplys E, Frederiksen JL, Houen G. Virus-specific antibody responses in severe acute respiratory syndrome coronavirus 2-infected and vaccinated individuals. Immunol Lett 2025; 274:107004. [PMID: 40157431 DOI: 10.1016/j.imlet.2025.107004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can have a serious course with many complications, especially in immunocompromised individuals. In such persons, other latent virus infections may contribute to disease pathology, in particular viruses which infect immune cells such as Epstein-Barr virus (EBV) and cytomegalovirus (CMV). METHODS In this study, serology-based assays were conducted to analyse antibody responses to SARS-CoV-2 spike protein (SP), EBV Epstein-Barr nuclear antigen (EBNA)-1 and CMV phosphoprotein (pp)52 in naturally SARS-CoV-2-infected individuals, non-infected healthy controls (HCs) and vaccinated healthy controls (VHCs) to identify an association between SARS-CoV-2 antibodies and EBV and CMV antibodies in order to determine whether latent EBV and CMV infected individuals are more prone to become infected with SARS-CoV-2. Moreover, SARS-CoV-2, EBV, and CMV antibody responses were characterized in serum from patients with relapsing-remitting multiple sclerosis (RRMS), a chronic inflammatory disease strongly associated with EBV infections, to determine whether the serologic virus antibody profile varies in immunocompromised RRMS individuals upon SARS-CoV-2 vaccinations compared to VHCs. RESULTS Significantly elevated SP IgG, IgM and IgA levels were identified in SARS-CoV-2-infected immunocompetent individuals when compared to non-infected HCs. However, no correlation was found to serum antibodies between SARS-CoV-2, EBV, and CMV in individuals infected with SARS-CoV-2 and in VHCs, suggesting that latent infections with neither EBV nor CMV associates to SARS-CoV-2 infection. Moreover, no significant difference in SP IgG, IgA and IgM levels was observed between vaccinated RRMS patients and VHCs, indicating that the immune system of immune deficient RRMS patients and VHCs respond identical to SARS-CoV-2 vaccinations. CONCLUSION Collectively, SARS-CoV-2 SP antibody levels reflect the vaccination and infection history and do not associate with EBV and CMV serostatus.
Collapse
Affiliation(s)
- Nicole Hartwig Trier
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark.
| | - Nadia Zivlaei
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark.
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, BLegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark.
| | - Margit Larsen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark.
| | - Rimantas Slibinskas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Evaldas Ciplys
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Jette Lautrup Frederiksen
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, BLegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Gunnar Houen
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55 5230 Odense M, Denmark.
| |
Collapse
|
9
|
Oba S, Hosoya T, Kawata D, Komiya Y, Iwai H, Koike R, Miyamoto S, Kanno T, Ainai A, Suzuki T, Hasegawa H, Yasuda S. Iguratimod, a promising therapeutic agent for COVID-19 that attenuates excessive inflammation in mouse models. Eur J Pharmacol 2025; 996:177537. [PMID: 40147575 DOI: 10.1016/j.ejphar.2025.177537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
In severe COVID-19 patients, excessive inflammation can lead to multiorgan dysfunction. Current anti-inflammatory treatments like glucocorticoids partially improve the outcomes, while immune systems are compromised. We have identified that SARS-CoV-2-infected obese mice were a good model of the cytokine storm seen in COVID-19. Here, we revealed that iguratimod (IGU), an approved agent for rheumatoid arthritis, improved survival by attenuating inflammation with minimal immune suppression. In this study, C57BL/6 mice were fed a high-fat diet (HFD) or a normal-fat diet (NFD) for ten weeks before being infected with a mouse-adapted SARS-CoV-2. IGU significantly improved survival rates and reduced lung inflammation in HFD-fed mice, with minimal impact on interferon-induced genes and viral load. Meanwhile, dexamethasone (DEX) did not improve survival, while it suppressed various immune reactions with different mechanisms to IGU. Interestingly, IGU-treated mice had fewer SARS-CoV-2 positive cells in the lung, although viral replication was comparable to the control mice. Neither IGU nor DEX inhibited the SARS-CoV-2 infection in Vero-E6 cells, unlike the antiviral agent, remdesivir. Of note, IGU was effective prophylactically and therapeutically in HFD mice, and showed beneficial effects in NFD-fed mice with a lethal dose exposure of SARS-CoV-2. We demonstrated that IGU could be a promising treatment for severe COVID-19, especially in obese patients, by fine-tuning inflammation without compromising antiviral immunity. This study supports the possibility of drug repositioning for IGU COVID-19 beyond autoimmune diseases.
Collapse
Affiliation(s)
- Seiya Oba
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Tadashi Hosoya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan.
| | - Daisuke Kawata
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Yoji Komiya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Hideyuki Iwai
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Ryuji Koike
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Takayuki Kanno
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Hideki Hasegawa
- WHO Collaborating Centre for Reference and Research on Influenza, Tokyo, Japan; Research Center for Influenza and Respiratory Virus, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan.
| |
Collapse
|
10
|
Anderson M, Lopez J, Wyr M, Ramirez PW. Defining diverse spike-receptor interactions involved in SARS-CoV-2 entry: Mechanisms and therapeutic opportunities. Virology 2025; 607:110507. [PMID: 40157321 DOI: 10.1016/j.virol.2025.110507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is an enveloped RNA virus that caused the Coronavirus Disease 2019 (COVID-19) pandemic. The SARS-CoV-2 Spike glycoprotein binds to angiotensin converting enzyme 2 (ACE2) on host cells to facilitate viral entry. However, the presence of SARS-CoV-2 in nearly all human organs - including those with little or no ACE2 expression - suggests the involvement of alternative receptors. Recent studies have identified several cellular proteins and molecules that influence SARS-CoV-2 entry through ACE2-dependent, ACE2-independent, or inhibitory mechanisms. In this review, we explore how these alternative receptors were identified, their expression patterns and roles in viral entry, and their impact on SARS-CoV-2 infection. Additionally, we discuss therapeutic strategies aimed at disrupting these virus-receptor interactions to mitigate COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Michael Anderson
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Julian Lopez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Maya Wyr
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Peter W Ramirez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Wu H, Liu Z, Li Y. Intestinal microbiota and respiratory system diseases: Relationships with three common respiratory virus infections. Microb Pathog 2025; 203:107500. [PMID: 40139334 DOI: 10.1016/j.micpath.2025.107500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
In recent years, the role of the intestinal microbiota in regulating host health and immune balance has attracted widespread attention. This study provides an in-depth analysis of the close relationship between the intestinal microbiota and respiratory system diseases, with a focus on three common respiratory virus infections, including respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and influenza virus. The research indicates that during RSV infection, there is a significant decrease in intestinal microbial diversity, suggesting the impact of the virus on the intestinal ecosystem. In SARS-CoV-2 infection, there are evident alterations in the intestinal microbiota, which are positively correlated with the severity of the disease. Similarly, influenza virus infection is associated with dysbiosis of the intestinal microbiota, and studies have shown that the application of specific probiotics exhibits beneficial effects against influenza virus infection. Further research indicates that the intestinal microbiota exerts a wide and profound impact on the occurrence and development of respiratory system diseases through various mechanisms, including modulation of the immune system and production of short-chain fatty acids (SCFAs). This article comprehensively analyzes these research advances, providing new perspectives and potential strategies for the prevention and treatment of future respiratory system diseases. This study not only deepens our understanding of the relationship between the intestinal microbiota and respiratory system diseases but also offers valuable insights for further exploring the role of host-microbiota interactions in the development of diseases.
Collapse
Affiliation(s)
- Haonan Wu
- Department of Respiratory, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China
| | - Ziyu Liu
- The First Hospital of Jilin University, Changchun, China.
| | - Yanan Li
- Department of Respiratory, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
14
|
Liao Y, Zeng T, Guo X, Li X. Ferritin's role in infectious diseases: Exploring pathogenic mechanisms and clinical implications. New Microbes New Infect 2025; 65:101582. [PMID: 40230813 PMCID: PMC11995792 DOI: 10.1016/j.nmni.2025.101582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Ferritin, an iron storage protein, is crucial for maintaining iron metabolism balance throughout the body and serves as a key biomarker for evaluating the body's iron reserves. Reduced ferritin levels typically indicate iron deficiency, whereas elevated ferritin levels indicate an acute inflammatory response in infectious diseases. Recent research has established a significant link between elevated ferritin levels and disease severity and prognosis. The concept of hyperferritinemic syndrome has underscored ferritin's role as a pathogenic mediator. During infections, ferritin not only serves as a biomarker of inflammation but also exerts pro-inflammatory functions, which is a key factor in perpetuating the vicious pathogenic cycle. This review offers a comprehensive exploration of ferritin, covering its structural characteristics, regulatory mechanisms, and how diverse pathogens modulate ferritin. Understanding its pivotal role in infectious diseases is essential for identifying novel therapeutic prospects and enhancing disease management and prevention.
Collapse
Affiliation(s)
| | | | - Xiaoyan Guo
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xinhua Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| |
Collapse
|
15
|
Sang JC, Musyoki SK, Injera WE, Karani LW, Maiyoh GK. Cytokine immune profiles among COVID 19 patients with different disease severities seeking treatment at Moi teaching and referral hospital, Kenya. Cytokine 2025; 190:156917. [PMID: 40117838 DOI: 10.1016/j.cyto.2025.156917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND COVID-19 manifests with a wide range of severities, from asymptomatic to critical conditions. Immunological profiles in patients positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may serve as early indicators of disease severity, aiding in prioritizing patient care. METHODOLOGY Archived patient plasma samples were retrieved from the Molecular Lab Bio-repository, ensuring equal representation of males, females, and various disease severities. Socio-demographic and disease severity data were obtained from patient health records. Levels of pro-inflammatory cytokines (interferon-gamma [IFN-γ], tumor necrosis factor-alpha [TNF-α], interleukin-2 [IL-2], and interleukin-17 [IL-17]) and anti-inflammatory cytokines (interleukin-4 [IL-4], interleukin-6 [IL-6], and interleukin-10 [IL-10]) were measured using the BD FACSCalibur flow cytometer. Data analysis involved comparing cytokine levels across different disease severities, with demographic data expressed as means ± standard deviation (SD). Statistical significance was set at P ≤ 0.05. FINDINGS The mean ages for males and females were 49.6 ± 22.7 and 48.4 ± 23.7, respectively. Mean ages for disease severity categories were 33 ± 19 (asymptomatic), 45.2 ± 21.5 (moderate), 56.8 ± 18.7 (severe), and 61.95 ± 22 (critical). Comorbidities were present in 25 % of patients, with cardiovascular disease (41 %) and pulmonary disease (31 %) being the most common. Predominant symptoms in critical patients included dyspnea (63 %) and myalgia (60 %), while rhinorrhea (46.2 %) and chest pain (45.7 %) were common in severe cases. Gastrointestinal symptoms were observed only in severe and critical groups. Levels of the pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-17) increased linearly with disease severity. Among anti-inflammatory cytokines, IL-6 and IL-10 levels also rose significantly with increasing severity. CONCLUSION Levels of TNF-α, IL-17, and IL-6 correlated with disease severity and may serve as prognostic biomarkers. Advanced age and underlying comorbidities were independently associated with higher disease severity.
Collapse
Affiliation(s)
- Jenniffer C Sang
- Department of Medical Laboratory Sciences, School of Health Sciences, Kisii University, Kisii, Kenya; Department of Laboratory and pathology Services, Moi Teaching and Referral Hospital, P.O. Box 3 - 30100, Eldoret, Kenya.
| | - Stanslaus K Musyoki
- Department of Medical Laboratory Sciences, School of Health Sciences, South Eastern Kenya University, P.O. Box 170-90200, Kitui, Kenya
| | - Wilfred E Injera
- Department of Medical Laboratory Sciences, School of Health Sciences, Alupe University, Busia, Kenya
| | - Lucy W Karani
- Department of Medical Laboratory Sciences, School of Health Sciences, Kisii University, Kisii, Kenya
| | - Geoffrey K Maiyoh
- Department of Biochemistry and Clinical Chemistry, School of Medicine, Moi University, P.O Box 4606-30100, Eldoret, Kenya.
| |
Collapse
|
16
|
Yasmin S, Ansari MY. A detailed examination of coronavirus disease 2019 (COVID-19): Covering past and future perspectives. Microb Pathog 2025; 203:107398. [PMID: 39986548 DOI: 10.1016/j.micpath.2025.107398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/07/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
The COVID-19 disease has spread rapidly across the world within just six months, affecting 169 million people and causing 3.5 million deaths globally (2021). The most affected countries include the USA, Brazil, India, and several European countries such as the UK and Russia. Healthcare professionals face new challenges in finding better ways to manage patients and save lives. In this regard, more comprehensive research is needed, including genomic and proteomic studies, personalized medicines and the design of suitable treatments. However, finding novel molecular entities (NME) using a standard or de novo strategy to drug development is a time-consuming and costly process. Another alternate strategy is discovering new therapeutic uses for old/existing/available medications, known as drug repurposing. There are a variety of computational repurposing methodologies, and some of them have been used to counter the coronavirus disease pandemic of 2019 (COVID-19). This review article compiles recently published data on the origin, transmission, pathogenesis, diagnosis, and management of the coronavirus by drug repurposing and vaccine development approach. We have attempted to screen probable drugs in clinical trials by using literature survey. This systematic review aims to create priorities for future research of drugs repurposed and vaccine development for COVID-19.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India; Ibne Seena College of Pharmacy, Azmi Vidya Nagri Anjhi Shahabad, Hardoi, Uttar Pradesh (U.P.) 241124, India.
| |
Collapse
|
17
|
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] [MESH Headings] [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
|
18
|
Zhang Y, Zhu F, Zhang Z, Wang J, Liao T, Xi Y, Liu D, Zhang H, Lin H, Mao J, Tang W, Zhao L, Yuan P, Yan L, Liu Q, Hong K, Qiao J. Alterations in Semen Quality and Immune-Related Factors in Men with Infertility who Recovered from COVID-19. MedComm (Beijing) 2025; 6:e70179. [PMID: 40276648 PMCID: PMC12019875 DOI: 10.1002/mco2.70179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
The emergence of coronavirus disease 2019 (COVID-19) has triggered research into its impact on male reproductive health. However, studies exploring the effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on semen quality in infertile men remain limited. Herein, we enrolled 781 male infertile patients who recovered from COVID-19 and analyzed their semen and blood samples collected at different time points. We found that SARS-CoV-2 RNA was undetectable in semen samples. Compared with pre-COVID-19 status, total sperm count, sperm concentration, vitality, motility, and percentage of sperm cells with normal morphology decreased significantly in the first month post-COVID-19. However, these alterations were reversed in the third month. Furthermore, seminal plasma samples exhibited reduced proinflammatory cytokine levels and notable changes in amino acid, nucleic acid, and carbohydrate metabolism by the third month compared with those in the first month. By contrast, no significant alterations in reproductive hormone levels were found. Vitality, progressive motility, and total motility negatively correlated with body temperature when it was above 38°C. In conclusion, semen quality initially decreases post-COVID-19 but reverses after approximately 3 months, with a decline related to inflammatory and fever. These findings may provide guidance to infertile male patients who need assisted reproductive technology.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Female Fertility PromotionNational Clinical Research Center for Obstetrics and GynecologyKey Laboratory of Assisted Reproduction (Peking University)Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyPeking University Third HospitalBeijingChina
| | - Feiyin Zhu
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Female Fertility PromotionNational Clinical Research Center for Obstetrics and GynecologyKey Laboratory of Assisted Reproduction (Peking University)Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyPeking University Third HospitalBeijingChina
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| | - Zhe Zhang
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Jing Wang
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Female Fertility PromotionNational Clinical Research Center for Obstetrics and GynecologyKey Laboratory of Assisted Reproduction (Peking University)Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyPeking University Third HospitalBeijingChina
| | - Tianyi Liao
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Female Fertility PromotionNational Clinical Research Center for Obstetrics and GynecologyKey Laboratory of Assisted Reproduction (Peking University)Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyPeking University Third HospitalBeijingChina
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| | - Yu Xi
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Defeng Liu
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Haitao Zhang
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Haocheng Lin
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Jiaming Mao
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Wenhao Tang
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Lianming Zhao
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Peng Yuan
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Female Fertility PromotionNational Clinical Research Center for Obstetrics and GynecologyKey Laboratory of Assisted Reproduction (Peking University)Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyPeking University Third HospitalBeijingChina
| | - Liying Yan
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Female Fertility PromotionNational Clinical Research Center for Obstetrics and GynecologyKey Laboratory of Assisted Reproduction (Peking University)Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyPeking University Third HospitalBeijingChina
| | - Qiang Liu
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Female Fertility PromotionNational Clinical Research Center for Obstetrics and GynecologyKey Laboratory of Assisted Reproduction (Peking University)Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyPeking University Third HospitalBeijingChina
| | - Kai Hong
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Jie Qiao
- Department of Obstetrics and GynecologyCenter for Reproductive MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Female Fertility PromotionNational Clinical Research Center for Obstetrics and GynecologyKey Laboratory of Assisted Reproduction (Peking University)Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive TechnologyPeking University Third HospitalBeijingChina
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
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
|
22
|
Vidal N, Climent MÁ, Pérez S, Méndez-Vidal MJ, Anguera G, Martínez Salas I, Gallardo E, Cuéllar-Rivas MA, Molina-Cerrillo J, Martín A, Rodriguez-Vida A, Almagro Casado E, Gonzalez M, Domènech M, Martínez Kareaga M, Fernández Calvo O, Villa Guzmán JC, Vázquez Estévez S, González-Del-Alba A, Puente J. Impact of COVID-19 infection on genitourinary cancer management. SOGUG-COVID-19: A spanish, multicenter, observational study. Clin Transl Oncol 2025; 27:2220-2231. [PMID: 39369361 DOI: 10.1007/s12094-024-03744-6] [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] [Accepted: 09/21/2024] [Indexed: 10/07/2024]
Abstract
INTRODUCTION The COVID-19 pandemic is a great burden worldwide, but its impact on patients with genitourinary cancer (GUC) is poorly characterized. This study aimed to characterize the clinical features and evolution of GUC patients affected by COVID-19 in Spain. PATIENTS AND METHODS SOGUG-COVID-19 was an observational ambispective non-interventional study that recruited patients with SARS-CoV-2 infection who had been treated for GUC in 32 Spanish hospitals. Data were collected from patients' medical records in a short period of time, coinciding with the first waves of COVID-19, when the mortality was also higher in the general population. RESULTS From November 2020 to April 2021, 408 patients were enrolled in the study. The median age was 70 years, and 357 patients (87.5%) were male. Most frequent Cancer Origin was: prostate (40.7%), urothelial (31.4%) and kidney (22.1%). Most patients (71.3%) were diagnosed at the metastatic stage, and 33.3% had poorly differentiated histology. Anticancer treatment during the infection was reported in 58.3% of patients, and 21.3% had received immunotherapy prior to or concurrent with the infection. The most frequent COVID-19 symptoms were pyrexia (49.0%), cough (38.2%) and dyspnea (31.9%). Median age was higher for patients with pneumonia (p < 0.001), patchy infiltrates (p = 0.005), ICU admission (p < 0.001) and death (p < 0.001). Tumor stage was associated with complications (p = 0.006). The fatality rate was 19.9% and the 6-month COVID-19-specific survival rate was 79.7%. CONCLUSION Patients with genitourinary cancers seem exceptionally vulnerable to COVID-19 regardless of tumor type or anticancer therapy. Age and tumor stage were the only identified risk factors for severe COVID-19.
Collapse
Affiliation(s)
- Natalia Vidal
- Medical Oncology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), CIBERONC, Madrid, Spain
| | | | - Sara Pérez
- Medical Oncology Department, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - María José Méndez-Vidal
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC) Hospital Universitario Reina Sofía, Medical Oncology Department, Córdoba, Spain
| | - Georgia Anguera
- Medical Oncology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Enrique Gallardo
- Medical Oncology Department, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Miler Andrés Cuéllar-Rivas
- Medical Oncology Department, Institut Català d'Oncologia (ICO) L'Hospitalet del Llobregat, Barcelona, Spain
| | | | - Almudena Martín
- Medical Oncology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Alejo Rodriguez-Vida
- Medical Oncology Department, Hospital del Mar, IMIM Research Institute, CIBERONC, Barcelona, Spain
| | - Elena Almagro Casado
- Medical Oncology Department, Hospital Universitario Quirón Salud Madrid, Pozuelo de Alarcón, Spain
| | - Macarena Gonzalez
- Medical Oncology Department, Vall d´Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d´Hebron, Barcelona, Spain
| | | | | | - Ovidio Fernández Calvo
- Medical Oncology Department, Complejo Hospitalario Universitario de Ourense, Ourense, Spain
| | | | | | - Aránzazu González-Del-Alba
- Medical Oncology Department, Hospital Universitario Puerta de Hierro-Majadahonda, C/Joaquin Rodrigo 2, Majadahonda, 28222, Madrid, Spain.
| | - Javier Puente
- Medical Oncology Department, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), CIBERONC, Madrid, Spain
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Rezaei Ghahroodi Z, Eftekhari Mahabadi S, Esberizi A, Sami R, Mansourian M. Association of the medication protocols and longitudinal change of COVID-19 symptoms: a hospital-based mixed-statistical methods study. J Biopharm Stat 2025; 35:386-406. [PMID: 38515283 DOI: 10.1080/10543406.2024.2333527] [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: 08/10/2023] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
The objective of this study was to identify the relationship between hospitalization treatment strategies leading to change in symptoms during 12-week follow-up among hospitalized patients during the COVID-19 outbreak. In this article, data from a prospective cohort study on COVID-19 patients admitted to Khorshid Hospital, Isfahan, Iran, from February 2020 to February 2021, were analyzed and reported. Patient characteristics, including socio-demographics, comorbidities, signs and symptoms, and treatments during hospitalization, were investigated. Also, to investigate the treatment effects adjusted by other confounding factors that lead to symptom change during follow-up, the binary classification trees, generalized linear mixed model, machine learning, and joint generalized estimating equation methods were applied. This research scrutinized the effects of various medications on COVID-19 patients in a prospective hospital-based cohort study, and found that heparin, methylprednisolone, ceftriaxone, and hydroxychloroquine were the most frequently prescribed medications. The results indicate that of patients under 65 years of age, 76% had a cough at the time of admission, while of patients with Cr levels of 1.1 or more, 80% had not lost weight at the time of admission. The results of fitted models showed that, during the follow-up, women are more likely to have shortness of breath (OR = 1.25; P-value: 0.039), fatigue (OR = 1.31; P-value: 0.013) and cough (OR = 1.29; P-value: 0.019) compared to men. Additionally, patients with symptoms of chest pain, fatigue and decreased appetite during admission are at a higher risk of experiencing fatigue during follow-up. Each day increase in the duration of ceftriaxone multiplies the odds of shortness of breath by 1.15 (P-value: 0.012). With each passing week, the odds of losing weight increase by 1.41 (P-value: 0.038), while the odds of shortness of breath and cough decrease by 0.84 (P-value: 0.005) and 0.56 (P-value: 0.000), respectively. In addition, each day increase in the duration of meropenem or methylprednisolone decreased the odds of weight loss at follow-up by 0.88 (P-value: 0.026) and 0.91 (P-value: 0.023), respectively (among those who took these medications). Identified prognostic factors can help clinicians and policymakers adapt management strategies for patients in any pandemic like COVID-19, which ultimately leads to better hospital decision-making and improved patient quality of life outcomes.
Collapse
Affiliation(s)
- Zahra Rezaei Ghahroodi
- School of Mathematics, Statistics and Computer Science, University of Tehran, Tehran, Iran
| | | | - Alireza Esberizi
- School of Mathematics, Statistics and Computer Science, University of Tehran, Tehran, Iran
| | - Ramin Sami
- Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marjan Mansourian
- Department of Epidemiology and Biostatistics, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
26
|
Agrawal A, Bajaj S, Bhagat U, Chandna S, Arockiam AD, Chan N, Haroun E, Gupta R, Badwan O, Shekhar S, Kathavarayan Ramu S, Nayar D, Jaber W, Griffin BP, Wang TKM. Intracardiac Thrombus in COVID-19 Inpatients: A Nationwide Study of Incidence, Predictors, and Outcomes. Angiology 2025; 76:441-452. [PMID: 38173053 DOI: 10.1177/00033197231225282] [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: 01/05/2024]
Abstract
COronaVIrus Disease-2019 (COVID-19) is associated with a hypercoagulable state. Intracardiac thrombosis is a potentially serious complication but has seldom been evaluated in COVID-19 patients. We assessed the incidence, associated factors, and outcomes of COVID-19 patients with intracardiac thrombosis. In 2020, COVID-19 inpatients were identified from the National Inpatient Sample (NIS) database. Data on clinical characteristics, intracardiac thrombosis, and adverse outcomes were collected. Multivariable logistic regression was used to identify factors associated with intracardiac thrombosis, in-hospital mortality, and morbidities. In 2020, 1,683,785 COVID-19 inpatients (mean age 63.8 years, 32.2% females) were studied. Intracardiac thrombosis occurred in 0.10% (1830) of cases. In-hospital outcomes included 13.2% all-cause mortality, 3.5% cardiovascular mortality, 2.6% cardiac arrest, 4.4% acute coronary syndrome (ACS), 16.1% heart failure, 1.3% stroke, and 28.3% acute kidney injury (AKI). Key factors for intracardiac thrombosis were congestive heart failure history and coagulopathy. Intracardiac thrombosis independently linked to higher risks of all-cause mortality (odds ratio [OR]: 3.32 (2.42-4.54)), cardiovascular mortality (OR: 2.95 (1.96-4.44)), cardiac arrest (OR: 2.04 (1.22-3.43)), ACS (OR: 1.62 (1.17-2.22)), stroke (OR: 3.10 (2.11-4.56)), and AKI (OR: 2.13 (1.68-2.69)), but not heart failure. While rare, intracardiac thrombosis in COVID-19 patients independently raised in-hospital mortality and morbidity risks.
Collapse
Affiliation(s)
- Ankit Agrawal
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suryansh Bajaj
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Umesh Bhagat
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Sanya Chandna
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Aro Daniela Arockiam
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nicholas Chan
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elio Haroun
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rahul Gupta
- Lehigh Valley Heart Institute, Lehigh Valley Health Network, Allentown, PA, USA
| | - Osamah Badwan
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shashank Shekhar
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shivabalan Kathavarayan Ramu
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Divya Nayar
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Wael Jaber
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brian P Griffin
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tom Kai Ming Wang
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
27
|
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] [MESH Headings] [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
|
28
|
Guo F, Wang Y, Chen J, Wang R, Wang L, Hong W, Du Y, Yang G. Construction and application of macrophage-based extracellular drug-loaded delivery systems. Int J Pharm 2025; 675:125462. [PMID: 40101875 DOI: 10.1016/j.ijpharm.2025.125462] [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: 09/19/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
Given their unique phagocytic function, inflammatory site tropism, and ability to penetrate deep into the lesion sites, macrophages are considered to have promising application potential in the field of living-cell drug delivery. The methods of drug delivery using macrophages primarily include intracellular phagocytic and extracellular drug loading. Comparatively, extracellular drug loading is potential less cytotoxicity and has minimal effects on the motility and orientation of cells. In this review, we provide an overview of the methods of extracellular drug loading, and examine the effects of the different properties of nanoformulations on extracellular drug-loaded delivery systems. In addition, we assess the prospects and challenges of a self-propelled macrophage-based drug delivery system. We hope this research contributes to optimizing the design of these drug delivery systems.
Collapse
Affiliation(s)
- Fangyuan Guo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yujia Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jialin Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Ruorong Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lianyi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Weiyong Hong
- Department of Pharmacy, Taizhou Municipal Hospital Affiliated to Taizhou University, Taizhou 318000, China
| | - Yinzhou Du
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
29
|
Ghaffarpour S, Ghazanfari T, Ardestani SK, Naghizadeh MM, Vaez Mahdavi MR, Salehi M, Majd AMM, Rashidi A, Chenary MR, Mostafazadeh A, Rezaei A, Khodadadi A, Iranparast S, Khazaei HA. Cytokine profiles dynamics in COVID-19 patients: a longitudinal analysis of disease severity and outcomes. Sci Rep 2025; 15:14209. [PMID: 40269030 PMCID: PMC12019550 DOI: 10.1038/s41598-025-98505-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/11/2025] [Indexed: 04/25/2025] Open
Abstract
The outcome of the immune response depends on the content and magnitude of inflammatory mediators, the right time to start, and the duration of inflammatory responses. Patients with coronavirus disease 2019 (COVID-19) represent diverse disease severity. Understanding differences in immune responses in individuals with different disease severity levels can help elucidate disease mechanisms. Here, we serially analyzed the cytokine profiles of 809 patients with mild to critical COVID-19. The cytokine profile revealed an overall increase in IL-1β, IL-1Ra, TNF-α, IL-6, IL-2, IL-8, and IL-18 and impaired production of IFN-α and -β. Only an early rise in IL-1Ra, IL-6, and IL-2 levels was linked to worse disease outcomes. On the other hand, long-term rises in IL-1β, IL-1Ra, TNF-α, IL-6, IL-2, IL-8, and IL-18 levels were linked to worse disease outcomes. Principal component analysis identified a component, including IL-1β, TNF-α, IFN-α, and IL-12, that was associated with disease severity. Spearman analysis revealed that the correlation of IL-1β and IFN-α was entirely different between mild and critical patients. Therefore, the ratio of IL-1β to IFN-α seemed to be a suitable criterion for distinguishing critical patients from mild ones. The higher levels of the IL-1β to IFN-α ratio correlated with improved outcomes. These data point to an imbalance of IL-1β/IFNα, contributing to hyperinflammation in COVID-19.
Collapse
Affiliation(s)
- Sara Ghaffarpour
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran.
- Department of Immunology, Shahed University, Tehran, Iran.
| | - Sussan Kaboudanian Ardestani
- Immunoregulation Research Center, Shahed University, Tehran, Iran
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | | | - Mohammadreza Salehi
- Department of Infectious Diseases, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Azadeh Rashidi
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | | | - Amrollah Mostafazadeh
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Khodadadi
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sara Iranparast
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Ali Khazaei
- Department of Immunology and Internal Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
30
|
Ali N, Hasan RA, Ibrahim IA, Mahmoud MF. Agomelatine attenuates Dexamethasone-Induced Neurotoxicity in Rats Through the activation of MT1/2 receptors and attenuation of oxidative stress. Eur J Pharmacol 2025:177659. [PMID: 40274180 DOI: 10.1016/j.ejphar.2025.177659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/10/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
Previous studies showed that agomelatine ameliorates doxorubicin-induced brain injury in rats. Furthermore, it protects neurons against oxidative stress triggered by acute ischemia reperfusion injury. So, this study aimed to investigate the possible neuroprotective effects of agomelatine on dexamethasone-induced neurotoxicity in rats and the underlying mechanisms. Subcutaneous injections of dexamethasone (10 mg/kg, 4 days) were used to induce neurotoxicity in rats. Agomelatine (10 mg/kg), luzindole (2.5 mg/kg, a melatonin receptor blocker), and luzindole plus agomelatine treatment commenced 3 days before dexamethasone injections and concurrent with dexamethasone injections. Elevated plus maze test, Y-maze test and open field test were carried out after 1 hour of the last dose of dexamethasone on day 7. On 8th day of the experiment, brain tissues were collected. Brain oxidative stress markers, immunohistochemical expression of β-amyloid and glial fibrillary acidic protein (GFAP) were measured. Moreover, histopathological changes in the cerebral cortex and hippocampus were recorded and the number of damaged cells was counted. Dexamethasone increased anxiety and memory impairment but decreased locomotor exploration activity. Furthermore, it increased brain oxidative stress, expression of β-amyloid and GFAP, increased the number of damaged neurons, and caused structural changes in cerebral cortex and hippocampus. All these deleterious changes were mitigated by agomelatine. Luzindole prior administration to agomelatine reversed the protective effects of agomelatine except its effect on lipid peroxidation. Collectively, these findings suggest that agomelatine can protect against dexamethasone-induced neurotoxicity partially by activating melatonin receptors in addition to exerting antioxidant effects.
Collapse
Affiliation(s)
- Noura Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| | - Rehab A Hasan
- Department of Histology, Faculty of Medicine for Girls, Al Azhar University, Cairo 11751, Egypt
| | - Islam Ahmed Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mona F Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
31
|
Barthez M, Xue B, Zheng J, Wang Y, Song Z, Mu WC, Wang CL, Guo J, Yang F, Ma Y, Wei X, Ye C, Sims N, Martinez-Sobrido L, Perlman S, Chen D. SIRT2 suppresses aging-associated cGAS activation and protects aged mice from severe COVID-19. Cell Rep 2025; 44:115562. [PMID: 40220296 DOI: 10.1016/j.celrep.2025.115562] [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: 04/06/2023] [Revised: 04/11/2024] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Aging-associated vulnerability to coronavirus disease 2019 (COVID-19) remains poorly understood. Here, we show that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected aged mice lacking SIRT2, a cytosolic NAD+-dependent deacetylase, develop more severe disease and show increased mortality, while treatment with an NAD+ booster, 78c, protects aged mice from lethal infection. Mechanistically, we demonstrate that SIRT2 modulates the acetylation of cyclic GMP-AMP synthase (cGAS), an immune sensor for cytosolic DNA, and suppresses aging-associated cGAS activation and inflammation. Furthermore, we show that SARS-CoV-2 infection-induced inflammation is mediated at least in part by ORF3a, which triggers mtDNA release and cGAS activation. Collectively, our study reveals a molecular basis for aging-associated susceptibility to COVID-19 and suggests therapeutic approaches to protect aged populations from severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Marine Barthez
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Biyun Xue
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Jian Zheng
- Department of Microbiology and Immunology, Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| | - Yifei Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Zehan Song
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Wei-Chieh Mu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chih-Ling Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jiayue Guo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Fanghan Yang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yuze Ma
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xuetong Wei
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Nicholas Sims
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Department of Pediatrics, University of Iowa, Iowa City, IA, USA.
| | - Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
32
|
Sohail MU, Aisha E, Waqas SA, Saad M, Arshad MS, Ahmed A, Sohail MO, Naveed Z, Amin E, Arora S, Jawaid H, Jain A, Memon MM. Trends in obesity-related ischemic heart disease mortality among adults in the United States from 1999 to 2020. Future Cardiol 2025:1-9. [PMID: 40255196 DOI: 10.1080/14796678.2025.2490397] [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: 11/07/2024] [Accepted: 04/04/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Obesity is a critical risk factor for ischemic heart disease (IHD), affecting 42% of the U.S. population. This study examines trends in obesity-related IHD mortality among U.S. adults aged 25 and older from 1999 to 2020, using the CDC WONDER database. RESEARCH DESIGN AND METHODS We analyzed IHD as the primary cause and obesity as a contributing factor, calculating age-adjusted (AAMRs) and crude mortality rates (CMRs) per 100,000 individuals. Joinpoint regression assessed annual percent changes (APC), stratifying by race, sex, age, and region. RESULTS From 1999 to 2020, 139,644 obesity-related IHD deaths were recorded. AAMR rose from 1.92 to 4.69 per 100,000. Rates were higher in men (3.79) than women (2.10), with Black Americans showing the highest AAMR (4.07). Older adults (65+) had the highest CMR (5.73). Nonmetropolitan areas exhibited higher AAMRs (3.47) than metropolitan regions (2.78). States with the highest mortality included Vermont, Oklahoma, Wyoming, Wisconsin and Iowa while Alabama, Virginia, Massachusetts, Connecticut and Georgia had the lowest. CONCLUSION The findings indicate a 2.5-fold increase in obesity-related IHD mortality, highlighting the need for targeted public health interventions and further research to address this growing public health concern.
Collapse
Affiliation(s)
| | - Eliza Aisha
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Saad Ahmed Waqas
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Muhammad Saad
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Aymen Ahmed
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Zara Naveed
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Emaan Amin
- Department Of Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Sahej Arora
- Department Of Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Hafsa Jawaid
- Department Of Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Aakriti Jain
- Department Of Medicine, Rochester General Hospital, Rochester, NY, USA
| | | |
Collapse
|
33
|
Sasankan S, Gathers D, Bellerose A, Pankratz VS, Tawfik B. Behaviors, perceptions, and impact of the COVID-19 pandemic and vaccination on oncology patients in New Mexico with substantial representation of racial minorities and rural residents. Vaccine 2025; 53:127091. [PMID: 40203593 DOI: 10.1016/j.vaccine.2025.127091] [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: 04/12/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
INTRODUCTION The COVID-19 pandemic has affected all sectors of life. This study helps us better understand the perceptions and practices related to the COVID-19 pandemic in the oncology patient population of New Mexico. It also explores patient knowledge regarding safe practices and perceptions regarding vaccination. METHODS The data for this cross-sectional survey study was collected from July 20, 2021 to September 6, 2021. RESULTS There were no significant differences noted in the incidence of COVID-19 infection based on race, comorbidities, or modality of treatment regardless of vaccination status. During the first peak of COVID-19 (Nov 2020 - Jan 2021), most participants followed strict safety precautions (54.2 %), fewer maintained these practices in the months prior to data collection (32.5 %) (Jul 2021 - Aug 2021). Among the participants who had declined vaccination against COVID-19, there were no significant differences based on race, comorbidities or treatment. Those oncology patients receiving treatments at the infusion center were much less vaccine hesitant (8.3 %) compared to those who were not (18.3 %). The odds of COVID-19 infection among patients that were vaccinated was 0.27 times lower than that of unvaccinated patients (95 % CI, 0.12 to 0.63; p 0.001). Vaccine-hesitant respondents reported long-term safety data (n = 11, 24.4 %) and physician recommendations (n = 10, 22.2 %) were likely to change their minds, but the most common response was that "nothing" would change their mind (n = 16, 35.6 %). CONCLUSION The pandemic is a dynamic landscape and physicians need to keep up to date with current guidelines and continue to have conversations with patients regarding strict safety precautions. Having an open discussion with patients regarding vaccine recommendations may help with decreasing vaccine hesitancy.
Collapse
Affiliation(s)
- S Sasankan
- Department of Internal Medicine, Division of Hematology and Oncology, Billings Clinic Cancer Center, 801 N 29th St, Billings, MT 59101, United States.
| | - D Gathers
- Department of Internal Medicine, Division of Hematology and Oncology, Duke University Health System, 2301 Erwin Road, Durham, NC 27710, United States.
| | - A Bellerose
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, United States.
| | - V S Pankratz
- Department of Internal Medicine, Division of Epidemiology, Biostatistics, and Preventative Medicine, University of New Mexico, 915 Camino de Salud NE, Albuquerque, Albuquerque, NM 87106, United States.
| | - B Tawfik
- Department of Internal Medicine, Division of Hematology and Oncology, University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM 87102, United States.
| |
Collapse
|
34
|
Vieira YP, da Silva LN, Nunes BP, Gonzalez TN, Duro SMS, de Oliveira Saes M. Relationship between long covid and functional disability in adults and the seniors in the south of Brazil. BMC Public Health 2025; 25:1458. [PMID: 40253365 PMCID: PMC12009519 DOI: 10.1186/s12889-025-22208-2] [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: 04/02/2024] [Accepted: 03/05/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Individuals living with long COVID experience a range of symptoms that affect their ability to carry out daily activities or participate in social and community life. This study aimed to analyze association between functional disability and the occurrence of long COVID symptoms, as well as to analyze the effect of symptom persistence time on functional disability. METHODS This is a cross-sectional study using data from the SulCovid-19 study, which interviewed individuals who had COVID-19 between December 2020 and March 2021. The functional disability outcome was assessed using the Basic Activities of Daily Living (BADL) and Instrumental Activities of Daily Living (IADL) scales, while the exposures were the symptoms of long COVID. Adjusted analyses between outcomes and exposures, stratified by time after the acute phase of infection, were performed using Poisson regression with robust variance adjustment. RESULTS The prevalence of BADL disability was 4.8% (95%CI 4.0;5.6), and for IADL disability, it was 8.4% (95%CI 7.4;9.4). The main symptoms associated with BADL disability were dyspnea, dry cough and sore throat, while for IADL, they were joint pain, muscle pain, loss of sensation, nasal congestion, sore throat and runny nose. When stratified by tertiles of time after the acute phase of infection, a relationship was found between BADL disability and dyspnea, ageusia and, nasal congestion in the 3rd tertile, while only ageusia was found to be related to IADL disability in the 3rd tertile. CONCLUSIONS Long COVID symptoms were associated wiht limitations in the functional capacity of adults and the seniors. The findings can be used to guide the care and rehabilitation of individuals with disabilities who have had COVID-19, particularly for referral to appropriate health professionals.
Collapse
Affiliation(s)
- Yohana Pereira Vieira
- Federal University of Rio Grande, rio Grande, Rio Grande, Rio Grande do Sul, Brazil.
- Institute/University/Hospital, Universidade Federal do Rio Grande, Country: Centro, St. General Osório, 102, 3º andar, Rio Grande, 96203-900, RS, Brasil.
| | | | | | | | | | | |
Collapse
|
35
|
Nouari W, Aribi M. Innate lymphoid cells, immune functional dynamics, epithelial parallels, and therapeutic frontiers in infections. Int Rev Immunol 2025:1-28. [PMID: 40242974 DOI: 10.1080/08830185.2025.2490233] [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: 04/03/2024] [Revised: 02/19/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Innate lymphoid cells (ILCs) have emerged as pivotal players in the field of immunology, expanding our understanding of innate immunity beyond conventional paradigms. This comprehensive review delves into the multifaceted world of ILCs, beginning with their serendipitous discovery and traversing their ontogeny and heterogeneity. We explore the distinct subsets of ILCs unraveling their intriguing plasticity, which adds a layer of complexity to their functional repertoire. As we journey through the functional activities of ILCs, we address their role in immune responses against various infections, categorizing their interactions with helminthic parasites, bacterial pathogens, fungal infections, and viral invaders. Notably, this review offers a detailed examination of ILCs in the context of specific infections, such as Mycobacterium tuberculosis, Citrobacter rodentium, Clostridium difficile, Salmonella typhimurium, Helicobacter pylori, Listeria monocytogenes, Staphylococcus aureus, Pseudomonas aeruginosa, Influenza virus, Cytomegalovirus, Herpes simplex virus, and severe acute respiratory syndrome coronavirus 2. This selection aimed for a comprehensive exploration of ILCs in various infectious contexts, opting for microorganisms based on extensive research findings rather than considerations of virulence or emergence. Furthermore, we raise intriguing questions about the potential immune functional resemblances between ILCs and epithelial cells, shedding light on their interconnectedness within the mucosal microenvironment. The review culminates in a critical assessment of the therapeutic prospects of targeting ILCs during infection, emphasizing their promise as novel immunotherapeutic targets. Nevertheless, due to their recent discovery and evolving understanding, effectively manipulating ILCs is challenging. Ensuring specificity and safety while evaluating long-term effects in clinical settings will be crucial.
Collapse
Affiliation(s)
- Wafa Nouari
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
36
|
Hatakeyama J, Nakamura K, Kanda N, Kawauchi A, Fujitani S, Oshima T, Kato H, Ota K, Kamijo H, Asahi T, Muto Y, Hori M, Iba A, Hosozawa M, Iso H. Long-term functional prognosis with tocilizumab in severe COVID-19 infection: A multicenter prospective observational study on mechanically ventilated ICU patients in the COVID-19 recovery study II. J Infect Chemother 2025; 31:102708. [PMID: 40250803 DOI: 10.1016/j.jiac.2025.102708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Tocilizumab, an IL-6 receptor antagonist, may prevent functional impairments in critically ill patients by attenuating the cytokine storm. This study investigated a potential effect of tocilizumab on preventing functional impairments in patients with severe coronavirus infection 2019 (COVID-19). METHODS In a multicenter prospective observational study, patients with COVID-19 ≥ 20 years requiring mechanical ventilation admitted to the intensive care unit between April 2021 and September 2021 and discharged alive were followed for one year. A self-administered questionnaire on sequelae and functional impairments was mailed in August 2022, and data were collected. A multivariate logistic regression was used to assess the impact of tocilizumab on physical function, mental health, and Long COVID. RESULTS Of 157 analyzed patients, 41 received tocilizumab. The tocilizumab group had more severe illness, but a lower prevalence of physical impairment (17.1 % vs. 23.3 %, p = 0.41) and mental disorders (19.5 % vs. 39.7 %, p = 0.009) than the non-tocilizumab group. The prevalence of Long COVID was higher in the tocilizumab group (92.7 % vs. 80.2 %, p = 0.06), whereas fatigue/malaise was significantly lower (19.5 % vs. 37.1 %, p = 0.039). Adjusted odds ratios (95 % confidence interval) for physical impairment, mental disorders, and Long COVID with tocilizumab were 0.70 (0.2-2.1), 0.40 (0.16-1.01), and 2.94 (0.7-12.3), respectively, with no significant difference. CONCLUSIONS Tocilizumab was associated with a lower prevalence of physical impairment and mental disorders at 1 year in patients with severe COVID-19. Furthermore, Long COVID had a weaker impact on physical and cognitive functions.
Collapse
Affiliation(s)
- Junji Hatakeyama
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kensuke Nakamura
- Department of Critical Care Medicine, Yokohama City University Hospital, Kanagawa, Japan; Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Ibaraki, Japan.
| | - Naoki Kanda
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Ibaraki, Japan; Division of General Internal Medicine, Jichi Medical University Hospital, Tochigi, Japan
| | - Akira Kawauchi
- Department of Critical Care and Emergency Medicine, Japanese Red Cross Maebashi Hospital, Gunma, Japan
| | - Shigeki Fujitani
- Department of Emergency Medicine and Critical Care Medicine, St. Marianna University, Kanagawa, Japan
| | - Taku Oshima
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, Kanagawa, Japan
| | - Kohei Ota
- Department of Emergency and Critical Care Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Hiroshi Kamijo
- Department of Emergency and Critical Care Medicine, Shinshu University School of Medicine, Nagano, Japan
| | - Tomohiro Asahi
- Department of Cardiology, Naha City Hospital, Okinawa, Japan
| | - Yoko Muto
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| | - Miyuki Hori
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| | - Arisa Iba
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| | - Mariko Hosozawa
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| | - Hiroyasu Iso
- Institute for Global Health Policy Research, Bureau of Global Health Cooperation, Japan Institute for Health Security, Tokyo, Japan
| |
Collapse
|
37
|
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
|
38
|
Sellarès-Nadal J, Espinosa-Pereiro J, Burgos J, Falcó V, Guillén-Del-Castillo A, Augustin S, Bañares-Sánchez J, Prio-Ruatg A, Martínez-Valle F, Kirkegaard-Biosca C, Sánchez-Montalvá A. Efficacy of tocilizumab for hospitalized patients with COVID-19 pneumonia and high IL-6 levels: A randomized controlled trial. Infection 2025:10.1007/s15010-025-02506-y. [PMID: 40232661 DOI: 10.1007/s15010-025-02506-y] [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: 11/20/2024] [Accepted: 03/04/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND The objective of this clinical trial is to evaluate the efficacy and safety of IL-6 driven personalized treatment strategy with tocilizumab in patients with severe COVID-19 pneumonia. TRIAL DESIGN Randomized, controlled, open-label, single-center trial of a tocilizumab treatment strategy in adult patients hospitalized with severe COVID-19 pneumonia and IL-6 serum levels > 40 pg/mL. METHODS Patients were randomized 1:1 to receive standard of care (SOC) or SOC plus one dose of tocilizumab. The primary outcome was death or need for invasive mechanical ventilation (IMV) within 28 days after randomization. Secondary outcomes included ICU admission, days on IMV and hospital stay. A meta-analysis of clinical trials to evaluate the effect of tocilizumab on mortality and need of IMV in patients with COVID-19 pneumonia was performed. RESULTS Sixty-two patients were included: 30 in the SOC arm and 32 in the standard-treatment plus tocilizumab arm. The primary outcome occurred in 12.9% in the tocilizumab arm and 32.3% in the SOC arm(p = 0.068). There was a trend towards fewer days on IMV (7.5 vs 19.5 days, p = 0.073) and a shorter hospital stay (4 vs 8 days, p = 0.134) in the tocilizumab group. No serious adverse events were reported. The meta-analysis revealed a RR for death or IMV of 0.83 (95% CI: 0.77-0.89) in patients receiving tocilizumab, compared to patients receiving SOC. CONCLUSION Tocilizumab could be effective to prevent death or IMV in patients with severe COVID-19 pneumonia and high IL-6 serum levels. Safety profile of tocilizumab does not arise major concern in patients with severe COVID19.
Collapse
Affiliation(s)
- Júlia Sellarès-Nadal
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Infectious Diseases Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Passeig Vall d'Hebrón 119-129, 08035, Barcelona, Spain
- Malalties Infeccioses Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Juan Espinosa-Pereiro
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Infectious Diseases Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Passeig Vall d'Hebrón 119-129, 08035, Barcelona, Spain
- Malalties Infeccioses Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Infectious Diseases Department, International Health Unit Vall d'Hebron-Drassanes, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquín Burgos
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Infectious Diseases Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Passeig Vall d'Hebrón 119-129, 08035, Barcelona, Spain.
- Malalties Infeccioses Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain.
| | - Vicenç Falcó
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Infectious Diseases Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Passeig Vall d'Hebrón 119-129, 08035, Barcelona, Spain
- Malalties Infeccioses Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Alfredo Guillén-Del-Castillo
- Internal Medicine Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Salvador Augustin
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Bañares-Sánchez
- Hepatology Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Alba Prio-Ruatg
- Hepatology Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Ferran Martínez-Valle
- Internal Medicine Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Cristina Kirkegaard-Biosca
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Infectious Diseases Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Passeig Vall d'Hebrón 119-129, 08035, Barcelona, Spain
- Malalties Infeccioses Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Adrián Sánchez-Montalvá
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Infectious Diseases Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Passeig Vall d'Hebrón 119-129, 08035, Barcelona, Spain
- Malalties Infeccioses Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Infectious Diseases Department, International Health Unit Vall d'Hebron-Drassanes, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
39
|
Isamail S, Idris IB, Mohd Yusoff H. Obstetric services preparedness among healthcare workers in maternal wards during COVID-19 pandemic. BMC Pregnancy Childbirth 2025; 25:443. [PMID: 40229684 PMCID: PMC11995598 DOI: 10.1186/s12884-025-07474-7] [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: 09/02/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic has been spreading rapidly since 2019, leading to devastating consequences worldwide, as well as putting healthcare providers at high risk. This study intends to assess the awareness and preparedness activities among maternal healthcare (MHC) workers and to determine factors that lead to obstetric services' preparedness in ten Government Hospitals in Selangor, Malaysia. METHODS A cross-sectional survey was conducted among 409 MHC workers which include doctors, nurses, and midwives in the government hospitals in Selangor, Malaysia between May 2022 till June 2022. Respondents were given validated questionnaires which include socio-demographic background, knowledge, awareness, and attitude as well as the obstetric services' preparedness activities in managing the COVID-19 pandemic. RESULTS Majority of the respondents were nurses/midwives (87.5%), female (98.5%), age 35-45 years old (48.5%), had working experience of more than 5 years (92.7%), had good knowledge and awareness (92.2%) and good obstetric services preparedness (88.3%). However, only about half (54.0%) of them had positive attitude towards COVID-19. When controlling for confounding factors, multivariate analysis showed that working period factor (p < 0.001), knowledge and awareness factor (p < 0.001) and attitude factor (p < 0.001) were significant predictive factors of obstetric services preparedness during the COVID-19 pandemic. CONCLUSIONS Majority of the respondents had good knowledge and awareness, as well as good level of obstetric services preparedness. However, only about half of them had a positive attitude towards COVID-19. The findings in this study revealed that efforts should be made to further increase the knowledge and attitude of MHC workers on COVID-19 and more so to further improve the positivity of their attitude towards this pandemic so that they can provide better obstetric services especially in the current and future pandemics to come.
Collapse
Affiliation(s)
- Sumaiyah Isamail
- Department of Public Health Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Idayu Badilla Idris
- Department of Public Health Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia.
| | - Hanizah Mohd Yusoff
- Department of Public Health Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| |
Collapse
|
40
|
Ge F, Tian F, Zhu Y, Yan Q, Sun Q, Lu J. Modified Xi-Jiao-Di-Huang Decoction Alleviates Sepsis via Regulating Macrophage Polarization by Inhibiting the PIM2/NF-κB Pathway. J Inflamm Res 2025; 18:5017-5030. [PMID: 40248592 PMCID: PMC12005212 DOI: 10.2147/jir.s509734] [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: 12/02/2024] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
Purpose Modified Xi-Jiao-Di-Huang decoction (MXJDH) has significant clinical efficacy for the treatment of sepsis; however, its mechanism of action remains unclear. The purpose of this study was to investigate the protective effects of MXJDH in septic mice and explore its mechanism of action. Methods Utilizing UPLC-Q-TOF-MS, we identified the primary constituents of the compound MXJDH. Subsequently, we created a mouse model for sepsis, observing their overall condition, including specific symptoms and behavior. We also monitored key inflammatory markers and pathological changes in their organs. Flow cytometry was then employed to assess the polarization of macrophages. Transcriptome sequencing was used to identify genes with altered expression patterns. We investigated the connection between MXJDH and the Pim2/NF-κB signaling pathway, a crucial regulatory mechanism in inflammation. Finally, we examined the expression and tissue distribution of macrophages in the sepsis-induced mice. Results MXJDH effectively reduces inflammation in sepsis mice, leading to a progressive recovery of organ functions. Moreover, MXJDH facilitates the conversion of macrophages from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotype. This transformation is potentially mediated through the Pim2/NF-κB signaling pathway. By suppressing Pim2 expression, MXJDH mitigates the nuclear translocation of NF-κB, thereby modulating the expression of downstream inflammatory mediators. The role of MXJDH in regulating macrophage polarization has also been confirmed in sepsis mouse tissues. Conclusion MXJDH regulates macrophage polarization, inhibits CRS, and alleviates sepsis by inhibiting the Pim2/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Fan Ge
- Department of Intensive Care Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Fang Tian
- Department of Central Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yeyan Zhu
- Department of Intensive Care Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Qixiang Yan
- Department of Intensive Care Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Qimeng Sun
- Department of Intensive Care Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Jun Lu
- Department of Intensive Care Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
41
|
Raspado O, Brack M, Brack O, Vivancos M, Esparcieux A, Cart-Tanneur E, Aouifi A. Oxidative Stress Markers and Prediction of Severity With a Machine Learning Approach in Hospitalized Patients With COVID-19 and Severe Lung Disease: Observational, Retrospective, Single-Center Feasibility Study. JMIR Form Res 2025; 9:e66509. [PMID: 40215478 PMCID: PMC12007842 DOI: 10.2196/66509] [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: 09/15/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 04/17/2025] Open
Abstract
Background Serious pulmonary pathologies of infectious, viral, or bacterial origin are accompanied by inflammation and an increase in oxidative stress (OS). In these situations, biological measurements of OS are technically difficult to obtain, and their results are difficult to interpret. OS assays that do not require complex preanalytical methods, as well as machine learning methods for improving interpretation of the results, would be very useful tools for medical and care teams. Objective We aimed to identify relevant OS biomarkers associated with the severity of hospitalized patients' condition and identify possible correlations between OS biomarkers and the clinical status of hospitalized patients with COVID-19 and severe lung disease at the time of hospital admission. Methods All adult patients hospitalized with COVID-19 at the Infirmerie Protestante (Lyon, France) from February 9, 2022, to May 18, 2022, were included, regardless of the care service they used, during the respiratory infectious COVID-19 epidemic. We collected serous biomarkers from the patients (zinc [Zn], copper [Cu], Cu/Zn ratio, selenium, uric acid, high-sensitivity C-reactive protein [hs-CRP], oxidized low-density lipoprotein, glutathione peroxidase, glutathione reductase, and thiols), as well as demographic variables and comorbidities. A support vector machine (SVM) model was used to predict the severity of the patients' condition based on the collected data as a training set. Results A total of 28 patients were included: 8 were asymptomatic at admission (grade 0), 14 had mild to moderate symptoms (grade 1) and 6 had severe to critical symptoms (grade 3). As the first outcome, we found that 3 biomarkers of OS were associated with severity (Zn, Cu/Zn ratio, and thiols), especially between grades 0 and 1 and between grades 0 and 2. As a second outcome, we found that the SVM model could predict the level of severity based on a biological analysis of the level of OS, with only 7% misclassification on the training dataset. As an illustrative example, we simulated 3 different biological profiles (named A, B, and C) and submitted them to the SVM model. Profile B had significantly high Zn, low hs-CRP, a low Cu/Zn ratio, and high thiols, corresponding to grade 0. Profile C had low Zn, low selenium, high oxidized low-density lipoprotein, high glutathione peroxidase, a low Cu/Zn ratio, and low glutathione reductase, corresponding to grade 2. Conclusions The level of severity of pulmonary damage in patients hospitalized with COVID-19 was predicted using an SVM model; moderate to severe symptoms in patients were associated with low Zn, low plasma thiol, increased hs-CRP, and an increased Cu/Zn ratio among a panel of 10 biomarkers of OS. Since this panel does not require a complex preanalytical method, it can be used and studied in other pathologies associated with OS, such as infectious pathologies or chronic diseases.
Collapse
Affiliation(s)
- Olivier Raspado
- Infirmerie Protestante, 1 Chemin du Penthod, Caluire-et-Cuire, 69300, France, 33 0624576962
| | - Michel Brack
- Oxidative Stress College, La Garenne-Colombes, France
| | - Olivier Brack
- Statistique Industrielle Khi² Consulting (KSIC), Bayet, France
| | - Mélanie Vivancos
- Clinical Research and Innovation Department, Infirmerie Protestante, Caluire-et-Cuire, France
| | - Aurélie Esparcieux
- Infirmerie Protestante, 1 Chemin du Penthod, Caluire-et-Cuire, 69300, France, 33 0624576962
| | | | - Abdellah Aouifi
- Infirmerie Protestante, 1 Chemin du Penthod, Caluire-et-Cuire, 69300, France, 33 0624576962
| |
Collapse
|
42
|
Igei T, Nakasone S, Takaesu M, Washizaki F, Kuniyoshi S, Onaga M, Otsuki K, Ishihara M, Yamakawa C, Nishida K. Characteristics of osteonecrosis of the femoral head after COVID-19 steroid treatment compared to steroid-associated osteonecrosis. BMC Musculoskelet Disord 2025; 26:352. [PMID: 40211205 PMCID: PMC11983807 DOI: 10.1186/s12891-025-08614-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Several studies have reported that the steroid dose of osteonecrosis of the femoral head (ONFH) after COVID-19 infection treatment was relatively low. However, the pathogenesis of ONFH after COVID-19 infection was not elucidated. This study aimed to analyze ONFH cases after COVID-19 infection and compare their characteristics with those of steroid-associated ONFH during the same period. METHODS This study includes 8 patients diagnosed with ONFH after steroid treatment for COVID-19 infection (COVID-19 group) from January 2020 to December 2023 in our institution and associated institutions. Nine patients diagnosed as steroid-associated ONFH without COVID-19 pneumonia treatment (SA-ONFH group) during same periods. Duration of steroid administration, cumulative dose of prednisolone equivalent, time to the onset of ONFH, history of alcohol and smoking, and blood test results were analyzed. Statistical analysis was performed between both groups using Mann-Whitney U test, Fisher's exact test (p < 0.05). RESULTS In COVID-19 group, the mean duration of steroid administration was 31 days, the mean cumulative dose of prednisolone equivalent was 719 mg, the mean time to the onset of ONFH was 9 months, 4 patients had a history of alcohol, 5 patients had a history of smoking. In SA-ONFH group, those were 681 days, 8,720 mg, 23 months, respectively; all patients have no alcohol and smoking history. In COVID-19 group, two patients had no history of alcohol and smoking. The administration duration of steroid in COVID-19 group was significantly shorter than that of SA-ONFH group. In addition, the mean cumulative dose of prednisolone equivalent was significantly lower than that of SA-ONFH group. A systematic inflammatory response and a hypercoagulable status could not be confirmed based on blood test results. CONCLUSIONS In our results, the mean cumulative dose in COVID-19 group was 719 mg, less than the diagnostic criteria (2000 mg) and our SA-ONFH group. Even in ONFH patients without history of alcohol and smoking in COVID-19 group, steroid dose were less than the criteria. Therefore COVID-19 infection itself or low-dose of steroid for COVID-19 patients may be a contributing factor to ONFH. Patients after COVID-19 infection treated with steroids should be followed for the possibility of developing ONFH.
Collapse
Affiliation(s)
- Takahiro Igei
- Department of orthopedic surgery, Graduate school of medicine, University of the Ryukyus, Kiyuna, Ginowan city, Okinawa, 1076, Japan
| | - Satoshi Nakasone
- Department of orthopedic surgery, Graduate school of medicine, University of the Ryukyus, Kiyuna, Ginowan city, Okinawa, 1076, Japan.
| | - Mika Takaesu
- Department of orthopedic surgery, Graduate school of medicine, University of the Ryukyus, Kiyuna, Ginowan city, Okinawa, 1076, Japan
| | - Fumiyuki Washizaki
- Department of orthopedic surgery, Graduate school of medicine, University of the Ryukyus, Kiyuna, Ginowan city, Okinawa, 1076, Japan
| | - Sakura Kuniyoshi
- Department of orthopedic surgery, Graduate school of medicine, University of the Ryukyus, Kiyuna, Ginowan city, Okinawa, 1076, Japan
| | - Masamichi Onaga
- Department of orthopedic surgery, Naha city hospital, 2-31-1, Furujima, Naha city, Okinawa, Japan
| | - Kenta Otsuki
- Department of orthopedic surgery, Nakagami hospital. 610, Noborikawa, Okinawa city, Okinawa, Japan
| | - Masato Ishihara
- Department of orthopedic surgery, Nakagami hospital. 610, Noborikawa, Okinawa city, Okinawa, Japan
| | - Chikashi Yamakawa
- Department of orthopedic surgery, Chubu Tokushukai hospital, 801, Higa, Kitanakagusuku-son, Okinawa, Japan
| | - Kotaro Nishida
- Department of orthopedic surgery, Graduate school of medicine, University of the Ryukyus, Kiyuna, Ginowan city, Okinawa, 1076, Japan
| |
Collapse
|
43
|
Sawant AN, Stensrud MJ. Explaining the sharp decline in birth rates in Canada and the United States in 2020. Am J Epidemiol 2025; 194:994-1001. [PMID: 39160447 PMCID: PMC11978608 DOI: 10.1093/aje/kwae274] [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: 11/23/2023] [Revised: 07/15/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024] Open
Abstract
Birth rates in Canada and the United States declined sharply in March 2020 and deviated from historical trends. This decline was absent in similarly developed European countries. We argue that the selective decline was driven by incoming individuals, who would have traveled from abroad and given birth in Canada and the United States had there been no travel restrictions during the COVID-19 pandemic. Furthermore, by leveraging data from periods before and during the COVID-19 travel restrictions, we quantified the extent of births by incoming individuals. In an interrupted time series analysis, the expected number of such births in Canada was 970 per month (95% CI, 710-1200), which is 3.2% of all births in the country. The corresponding estimate for the United States was 6700 per month (95% CI, 3400-10 000), which is 2.2% of all births. A secondary difference-in-differences analysis gave similar estimates, at 2.8% and 3.4% for Canada and the United States, respectively. Our study reveals the extent of births by recent international arrivals, which hitherto has been unknown and infeasible to study.
Collapse
Affiliation(s)
- Amit N Sawant
- Institute of Mathematics Ecole Polytechnique Fédérale de Lausanne, Lausanne, Vaud, Switzerland
| | - Mats J Stensrud
- Institute of Mathematics Ecole Polytechnique Fédérale de Lausanne, Lausanne, Vaud, Switzerland
| |
Collapse
|
44
|
Murakami T, Yamaguchi Y, Amiya S, Yoshimine Y, Nameki S, Okita Y, Kato Y, Hirata H, Takeda Y, Kumanogoh A, Morita T. CD147-high classical monocytes: a cellular biomarker for COVID-19 disease severity and treatment response. Inflamm Regen 2025; 45:8. [PMID: 40189583 PMCID: PMC11974131 DOI: 10.1186/s41232-025-00371-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can lead to severe coronavirus disease 2019 (COVID-19), which is characterized by cytokine storm and organ dysfunction. The spike S1 subunit induces inflammatory cytokine production, but the immune cell subsets that respond to S1 stimulation and contribute to disease severity remain unclear. METHODS We analyzed serum samples and peripheral blood mononuclear cells (PBMCs) from patients with COVID-19 (moderate: n = 7; severe: n = 25) and healthy controls (n = 38). Using mass cytometry (cytometry by time-of-flight; CyTOF), we analyzed immune cell responses to S1 subunit stimulation in PBMCs from healthy donors and patients with COVID-19. We examined correlations among identified cell populations, serum cytokine levels, and clinical parameters. RESULTS Serum S1 subunit levels correlated with disease severity and inflammatory cytokine concentrations. S1 subunit stimulation induced dose-dependent cytokine production from PBMCs, predominantly from myeloid cells. CyTOF analysis identified classical monocytes with high CD147 expression (CD147hi cMono) as the primary source of S1-induced cytokines. The proportion of CD147hi cMono increased significantly in severe COVID-19 and decreased with clinical improvement. The frequency of CD147hi cMono showed a stronger positive correlation with clinical severity markers in younger patients compared to older patients. CONCLUSIONS CD147hi cMono are the primary cellular source of S1-induced inflammatory cytokines and may serve as potential biomarkers for monitoring COVID-19 severity and treatment response.
Collapse
Affiliation(s)
- Teruaki Murakami
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Yuta Yamaguchi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Saori Amiya
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Yuko Yoshimine
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Shinichiro Nameki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Yasutaka Okita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Medical Center for Translational Research, Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, 565-0871, Japan
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan.
- Strategic Global Partnership & X(Cross)-Innovation Initiative, Graduate School of Medicine, Osaka University and Osaka University Hospital, Osaka, Japan.
| |
Collapse
|
45
|
Song Y, Wang X, Tong D, Huang X, Jin X, Zhang C, Liu J, Guo B, Huang C, Lian J. Identification of Potential Biomarkers and Immune Cell Signatures in COVID-19 Myocarditis Through Bioinformatic Analysis. Cardiol Res Pract 2025; 2025:2349610. [PMID: 40230577 PMCID: PMC11996287 DOI: 10.1155/crp/2349610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/27/2024] [Accepted: 03/01/2025] [Indexed: 04/16/2025] Open
Abstract
Objective: The present study aims to elucidate the significance of immune cell infiltration in Coronavirus disease 2019 (COVID-19) myocarditis and identify potential diagnostic markers for this condition. Myocarditis, an inflammatory cardiac disease, primarily results from viral infections. Although the association between COVID-19 and myocarditis is well-established, the specific mechanism(s) underlying this relationship remain incompletely understood. Methods: The GSE53607 and GSE35182 datasets were obtained from the GEO database, which contains samples from a mouse model for viral myocarditis. Differentially expressed genes (DEGs) and candidate biomarkers were selected using the LASSO regression model and support vector machine recursive feature elimination (SVM-RFE) analysis. Subsequently, the diagnostic potential of these biomarkers was evaluated by calculating the area under the receiver operating characteristic curve (AUC). Further validation of the biomarkers was conducted using the GSE183850 dataset, which consists of samples from patients with COVID-19 myocarditis. In addition, CIBERSORT analysis was employed to estimate the compositional patterns of 22 types of immune cell fractions in merged cohorts. Results: Thirty genes were identified, with a significant proportion of the DEGs being associated with carbohydrate binding, endopeptidase activity, and pathogenic organisms such as Staphylococcus aureus and coronavirus disease. Importantly, gene sets related to the IL6-JAK-STAT3 signaling pathways, inflammatory response, and interferon response exhibited differential activation in viral myocarditis compared to the control group. In addition, in the context of COVID-19 myocarditis patients from the GSE183850 dataset, B2M and C3 were established as diagnostic markers that were subsequently validated (AUC = 0.978 and AUC = 0.956, respectively). Furthermore, analysis of immune cell infiltration revealed correlations between B2M and C3 expression levels and the activation of NK cells, dendritic cells, T cells CD4 memory resting, as well as eosinophils. Conclusion: B2M and C3 have been identified as potential biomarkers for viral myocarditis, providing valuable insights for future investigations into the pathogenesis of COVID-19-associated myocarditis.
Collapse
Affiliation(s)
- Yongfei Song
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Xiaofei Wang
- Biomedical Experimental Center, Xi'an Jiaotong University, Xian, Shaanxi, China
| | - Dongdong Tong
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xian, Shaanxi, China
- Key Laboratory of Environmentally and Genetically Associated Diseases, Xi'an Jiaotong University, Ministry of Education, Xian, Shaanxi, China
| | - Xiaoyan Huang
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Xiaojun Jin
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Chuanjing Zhang
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Jianhui Liu
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Bo Guo
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xian, Shaanxi, China
- Key Laboratory of Environmentally and Genetically Associated Diseases, Xi'an Jiaotong University, Ministry of Education, Xian, Shaanxi, China
| | - Chen Huang
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Biomedical Experimental Center, Xi'an Jiaotong University, Xian, Shaanxi, China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xian, Shaanxi, China
- Key Laboratory of Environmentally and Genetically Associated Diseases, Xi'an Jiaotong University, Ministry of Education, Xian, Shaanxi, China
| | - Jiangfang Lian
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
46
|
Tian Y, Cipollo JF. Comparison of N- and O-Glycosylation on Spike Glycoprotein 1 of SARS-CoV-1 and MERS-CoV. J Proteome Res 2025. [PMID: 40193531 DOI: 10.1021/acs.jproteome.4c00716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
SARS-CoV-1 and MERS-CoV were the infective agents of the 2002 and 2012 coronavirus outbreaks, respectively. Here, we report a comparative liquid chromatography/mass spectrometry (LC/MS) Orbitrap N- and O-glycosylation glycoproteomics study of the recombinant S1 spike derived from these two viruses. The former was produced in HEK293 cells and the latter in both HEK293 and insect cells. Both proteins were highly glycosylated, with SARS-CoV-1 S1 having 13 and MERS-CoV S1 having 12 N-glycosites. Nearly all were occupied at 85% or more. Between 2 and 113 unique N-glycan compositions were detected at each N-glycosite across the three proteins. Complex N-glycans dominated in HEK293 cell-derived spike S1 proteins. While glycosylation differs between HEK293 and insect cells, the extent of glycan processing at glycosites was similar for the two MERS-CoV S1 forms. The HEK293-derived SARS-CoV-1 S1 N-glycans were more highly sialylated and fucosylated compared to MERS S1, while the latter had more high-mannose glycosides, particularly in the N-terminus and near the RBD. Seven and 8 O-glycosites were identified in SARS-CoV-1 S1 and MERS-CoV S1, respectively. Mapping of predicted antigenic and glycosylation sites reveals colocalization consistent with a role for glycosylation in immune system avoidance. Glycosylation patterns of these S1 proteins differ from those of other SARS-CoV-1 and MERS-CoV spike reported forms such as recombinant trimeric and virus-propagated forms, which has implications for virus research, including vaccine development, as glycosylation plays a role in spike function and epitope structure.
Collapse
Affiliation(s)
- Yuan Tian
- Food and Drug Administration, Center for Biologics Evaluation and Research, Division of Product Quality Assessment V, Parasitic and Allergenic Products, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States
- Food and Drug Administration, Center for Drug Evaluation and Research, Division of Product Quality Assessment, Parasitic and Allergenic Products, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States
| | - John F Cipollo
- Food and Drug Administration, Center for Biologics Evaluation and Research, Division of Product Quality Assessment V, Parasitic and Allergenic Products, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States
| |
Collapse
|
47
|
Marchesi N, Allegri M, Bruno GM, Pascale A, Govoni S. Exploring the Potential of Dietary Supplements to Alleviate Pain Due to Long COVID. Nutrients 2025; 17:1287. [PMID: 40219044 PMCID: PMC11990457 DOI: 10.3390/nu17071287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Long COVID, characterized by persistent symptoms following COVID-19 infection, significantly impacts individuals' health and daily functioning due to fatigue and pain. Focusing on pain, this review addresses nociplastic and chronic pain conditions. Interventions designed to reduce inflammation, oxidative stress, and enhance vagal activity may offer a promising approach to managing post-pandemic pain. This review presents individual components of food supplements with demonstrated efficacy in one or more pain conditions, focusing on their proposed mechanisms and clinical activity in pain, including their use in post-COVID-19 pain when available. Many of these substances have a long history of safe use and may offer an alternative to long-term analgesic drug treatment, which is often associated with potential side effects. This review also explores the potential for synergistic effects when combining these substances with each other or with conventional analgesics, considering the advantages for both patients and the healthcare system in using these substances as adjunctive or primary therapies for pain symptoms related to long COVID. While preclinical scientific literature provides a mechanistic basis for the action of several food supplements on pain control mechanisms and signaling pathways, clinical experience, particularly in the field of long COVID-associated pain, is still limited. However, the reviewed literature strongly suggests that the use of food supplements in long COVID-associated pain is an attainable goal, provided that rigorous clinical trials are conducted.
Collapse
Affiliation(s)
- Nicoletta Marchesi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, 27100 Pavia, Italy; (G.M.B.); (A.P.); (S.G.)
- RedyNeuheart s.r.l., Start-Up, Via Santa Marta 19, 20123 Milan, Italy
| | - Massimo Allegri
- Centre Lémanique de Neuromodulation et Thérapie de la Douleur, Hôpital de Morges, Ensemble Hospitalier de la Côte (EHC), 1110 Morges, Switzerland;
| | - Giacomo Matteo Bruno
- Department of Drug Sciences, Pharmacology Section, University of Pavia, 27100 Pavia, Italy; (G.M.B.); (A.P.); (S.G.)
- Center of Research, SAVE Studi—Health Economics and Outcomes Research, 20123 Milan, Italy
- CEFAT (Center of Pharmaceuticals Economics and Medical Technologies Evaluation), University of Pavia, 27100 Pavia, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Pharmacology Section, University of Pavia, 27100 Pavia, Italy; (G.M.B.); (A.P.); (S.G.)
| | - Stefano Govoni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, 27100 Pavia, Italy; (G.M.B.); (A.P.); (S.G.)
- CEFAT (Center of Pharmaceuticals Economics and Medical Technologies Evaluation), University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
48
|
Li X, Tian Y, Cao H, Cheng J. Serum sST2: key biomarkers in COVID-19 patients with implications for coronary artery disease. BMC Infect Dis 2025; 25:471. [PMID: 40197291 PMCID: PMC11974224 DOI: 10.1186/s12879-025-10849-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: 06/04/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND As the coronavirus disease-2019 (COVID-19) pandemic persists, post-COVID-19 syndrome (PS), characterized by symptoms like chest pain, fatigue, and palpitations, is becoming a significant medical and social issue. COVID-19 patients with existing coronary artery disease (CAD) may face higher risks of complications. It is crucial to assess if PS patients also have CAD, though data is limited. METHODS We studied 75 COVID-19 patients and 68 non-COVID-19 patients admitted to our hospital between 2022/12/20 to 2023/01/20. Demographic, laboratory, and clinical data were collected upon admission. The Gensini score (GS) was used to assess coronary atherosclerosis severity. Patients were categorized by GS and clinical traits to identify potential independent risks linked to CAD and COVID-19 severity. RESULTS COVID-19 patients with existing CAD had higher levels of serum soluble growth stimulation expression of gene 2 protein (sST2), myeloperoxidase, ALT, AST, PT, B-type natriuretic peptide (BNP), and hypersensitive troponin-I (hs-cTnI), along with longer hospital stays, more ICU admissions, and increased heart failure and ACS morbidity compared to those without CAD. Univariate and multivariate analysis identified sST2 as an independent risk factor for COVID-19 patients with coexisting CAD (odds ratio 1.122). sST2 levels were positively correlated with coronary angiography GS (r = 0.474, p < 0.001) in COVID-19 patients and were significantly higher in cases with GS ≥ 32, regardless of COVID-19 status (p < 0.001) and specifically in COVID-19 patients (p = 0.006). ROC analysis showed sST2 predicted ICU admission, hospital stay duration, and morbidity of HF and ACS similarly to GS. CONCLUSIONS Admission serum sST2 levels should be considered in COVID-19 patients with CAD-like symptoms for treatment planning and could serve as a prognostic biomarker for COVID-19 with co-existing CAD in clinical practice.
Collapse
Affiliation(s)
- Xueqin Li
- Department of Laboratory Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yaxin Tian
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
- Department of Health Statistics, Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Hongyan Cao
- Department of Health Statistics, Shanxi Provincial Key Laboratory of Major Diseases Risk Assessment, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Jinfang Cheng
- Department of Cardiovascular Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyaun, 030032, China.
| |
Collapse
|
49
|
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
|
50
|
Qin J, Wang G, Han D. Benefits of melatonin on mortality in severe-to-critical COVID-19 patients: A systematic review and meta-analysis of randomized controlled trials. Clinics (Sao Paulo) 2025; 80:100638. [PMID: 40187234 PMCID: PMC12002743 DOI: 10.1016/j.clinsp.2025.100638] [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: 10/02/2024] [Revised: 03/05/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE This meta-analysis aimed to determine the efficacy of melatonin on mortality in patients with severe-to-critical illness COVID-19. METHODS A systematic search was made of PubMed, Embase, Cochrane Library, and clinicaltrials.gov, without language restrictions. Randomized Controlled Trials (RCTs) on the treatment of severe-to-critical COVID-19 with melatonin, compared with placebo or blank, were reviewed. Studies were pooled to Odds Ratios (ORs), with 95 % Confidence Intervals (95 % CIs). RESULTS Three RCTs (enrolling 451 participants) met the inclusion criteria. Melatonin showed a significant effect on in-hospital mortality (OR = 0.19, 95 % CI 0.05 to 0.74; p = 0.02). CONCLUSIONS Melatonin significantly reduced in-hospital mortality in patients with severe-to-critical COVID-19. Melatonin should be considered for severe-to-critical COVID-19 patients.
Collapse
Affiliation(s)
- Jinlv Qin
- Radioimmunoassay Center, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Guizuo Wang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Dong Han
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, China.
| |
Collapse
|